Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
1.
Sci Rep ; 10(1): 20530, 2020 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-33239756

RESUMO

Enterotoxigenic Escherichia coli (ETEC), an essential cause of post-weaning diarrhea (PWD) in piglets, leads to significant economic losses to the pig industry. The present study aims to identify the role of ETEC total RNA in eliciting immune responses to protect animals against ETEC infection. The results showed that the total RNA isolated from pig-derived ETEC K88ac strain effectively stimulated the IL-1ß secretion of porcine intestinal epithelial cells (IPEC-J2). The mouse model immunized with ETEC total RNA via intramuscular injection (IM) or oral route (OR) was used to evaluate the protective efficiency of the ETEC total RNA. The results suggested that 70 µg ETEC total RNA administered by either route significantly promoted the production of the serum IL-1ß and K88ac specific immunoglobulins (IgG, IgM, and IgA). Besides, the ETEC RNA administration augmented strong mucosal immunity by elevating K88ac specific IgA level in the intestinal fluid. Intramuscularly administered RNA induced a Th1/Th2 shift toward a Th2 response, while the orally administered RNA did not. The ETEC total RNA efficiently protected the animals against the ETEC challenge either by itself or as an adjuvant. The histology characterization of the small intestines also suggested the ETEC RNA administration protected the small intestinal structure against the ETEC infection. Particularly of note was that the immunity level and protective efficacy caused by ETEC RNA were dose-dependent. These findings will help understand the role of bacterial RNA in eliciting immune responses, and benefit the development of RNA-based vaccines or adjuvants.


Assuntos
Escherichia coli Enterotoxigênica/imunologia , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/prevenção & controle , RNA Bacteriano/imunologia , Animais , Linhagem Celular , Citocinas/sangue , Citocinas/metabolismo , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Infecções por Escherichia coli/sangue , Infecções por Escherichia coli/microbiologia , Imunidade nas Mucosas , Imunização , Intestinos/patologia , Camundongos , Análise de Sobrevida , Suínos , Células Th1/metabolismo , Células Th2/metabolismo
2.
Sci Rep ; 10(1): 18293, 2020 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-33106559

RESUMO

Bacterial-derived RNA and DNA can function as ligands for intracellular receptor activation and induce downstream signaling to modulate the host response to bacterial infection. The mechanisms underlying the secretion of immunomodulatory RNA and DNA by pathogens such as Staphylococcus aureus and their delivery to intracellular host cell receptors are not well understood. Recently, extracellular membrane vesicle (MV) production has been proposed as a general secretion mechanism that could facilitate the delivery of functional bacterial nucleic acids into host cells. S. aureus produce membrane-bound, spherical, nano-sized, MVs packaged with a select array of bioactive macromolecules and they have been shown to play important roles in bacterial virulence and in immune modulation through the transmission of biologic signals to host cells. Here we show that S. aureus secretes RNA and DNA molecules that are mostly protected from degradation by their association with MVs. Importantly, we demonstrate that MVs can be delivered into cultured macrophage cells and subsequently stimulate a potent IFN-ß response in recipient cells via activation of endosomal Toll-like receptors. These findings advance our understanding of the mechanisms by which bacterial nucleic acids traffic extracellularly to trigger the modulation of host immune responses.


Assuntos
DNA Bacteriano/imunologia , Vesículas Extracelulares/genética , Macrófagos/virologia , RNA Bacteriano/imunologia , Staphylococcus aureus/patogenicidade , Animais , Vesículas Extracelulares/imunologia , Interferon gama/genética , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Tamanho da Partícula , Células RAW 264.7 , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia , Receptores Toll-Like/genética , Virulência
3.
BMC Microbiol ; 20(1): 217, 2020 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-32689952

RESUMO

BACKGROUND: Lactobacillus gasseri OLL2809 can highly induce interleukin (IL)-12 production in immune cells. Even though beneficial properties of this strain for both humans and animals have been reported, the mechanism by which the bacteria induces the production of IL-12 in immune cells remains elusive. In this study, we investigated the mechanism of induction of IL-12 using a mouse macrophage cell line J774.1. RESULTS: Inhibition of phagocytosis of L. gasseri OLL2809, and myeloid differentiation factor 88 and Toll-like receptors (TLRs) 7 and 9 signalling attenuated IL-12 production in J774.1 cells. Total RNA and genomic DNA of L. gasseri OLL2809, when transferred to the J774.1 cells, also induced IL-12 production. The difference in the IL-12-inducing activity of Lactobacilli is attributed to the susceptibility to phagocytosis, but not to a difference in the total RNA and genomic DNA of each strain. CONCLUSION: We concluded that total RNA and genomic DNA of phagocytosed L. gasseri OLL2809 induce IL-12 production in J774.1 cell via TLRs 7 and 9, and the high IL-12-inducing activity of L. gasseri OLL2809 is due to its greater susceptibility to phagocytosis.


Assuntos
DNA Bacteriano/imunologia , Interleucina-12/metabolismo , Lactobacillus gasseri/genética , Macrófagos/imunologia , Glicoproteínas de Membrana/metabolismo , RNA Bacteriano/imunologia , Receptor 7 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo , Animais , Técnicas de Cultura de Células , Linhagem Celular , Genoma Bacteriano , Lactobacillus gasseri/imunologia , Macrófagos/citologia , Macrófagos/microbiologia , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Fagocitose , Transdução de Sinais , Regulação para Cima
4.
J Neuroinflammation ; 17(1): 139, 2020 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32357908

RESUMO

BACKGROUND: Bacterial meningitis and meningoencephalitis are associated with devastating neuroinflammation. We and others have demonstrated the importance of glial cells in the initiation of immune responses to pathogens invading the central nervous system (CNS). These cells use a variety of pattern recognition receptors (PRRs) to identify common pathogen motifs and the cytosolic sensor retinoic acid inducible gene-1 (RIG-I) is known to serve as a viral PRR and initiator of interferon (IFN) responses. Intriguingly, recent evidence indicates that RIG-I also has an important role in the detection of bacterial nucleic acids, but such a role has not been investigated in glia. METHODS: In this study, we have assessed whether primary or immortalized human and murine glia express RIG-I either constitutively or following stimulation with bacteria or their products by immunoblot analysis. We have used capture ELISAs and immunoblot analysis to assess human microglial interferon regulatory factor 3 (IRF3) activation and IFN production elicited by bacterial nucleic acids and novel engineered nucleic acid nanoparticles. Furthermore, we have utilized a pharmacological inhibitor of RIG-I signaling and siRNA-mediated knockdown approaches to assess the relative importance of RIG-I in such responses. RESULTS: We demonstrate that RIG-I is constitutively expressed by human and murine microglia and astrocytes, and is elevated following bacterial infection in a pathogen and cell type-specific manner. Additionally, surface and cytosolic PRR ligands are also sufficient to enhance RIG-I expression. Importantly, our data demonstrate that bacterial RNA and DNA both trigger RIG-I-dependent IRF3 phosphorylation and subsequent type I IFN production in human microglia. This ability has been confirmed using our nucleic acid nanoparticles where we demonstrate that both RNA- and DNA-based nanoparticles can stimulate RIG-I-dependent IFN responses in these cells. CONCLUSIONS: The constitutive and bacteria-induced expression of RIG-I by human glia and its ability to mediate IFN responses to bacterial RNA and DNA and nucleic acid nanoparticles raises the intriguing possibility that RIG-I may be a potential target for therapeutic intervention during bacterial infections of the CNS, and that the use of engineered nucleic acid nanoparticles that engage this sensor might be a method to achieve this goal.


Assuntos
DNA Bacteriano/imunologia , Microglia/imunologia , RNA Bacteriano/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Receptores do Ácido Retinoico/imunologia , Animais , Células Cultivadas , Humanos , Fator Regulador 3 de Interferon/biossíntese , Interferons/biossíntese , Camundongos , Camundongos Endogâmicos C57BL
5.
Immunity ; 52(4): 591-605.e6, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32294405

RESUMO

Human toll-like receptor 8 (TLR8) activation induces a potent T helper-1 (Th1) cell response critical for defense against intracellular pathogens, including protozoa. The receptor harbors two distinct binding sites, uridine and di- and/or trinucleotides, but the RNases upstream of TLR8 remain poorly characterized. We identified two endolysosomal endoribonucleases, RNase T2 and RNase 2, that act synergistically to release uridine from oligoribonucleotides. RNase T2 cleaves preferentially before, and RNase 2 after, uridines. Live bacteria, P. falciparum-infected red blood cells, purified pathogen RNA, and synthetic oligoribonucleotides all required RNase 2 and T2 processing to activate TLR8. Uridine supplementation restored RNA recognition in RNASE2-/- or RNASET2-/- but not RNASE2-/-RNASET2-/- cells. Primary immune cells from RNase T2-hypomorphic patients lacked a response to bacterial RNA but responded robustly to small-molecule TLR8 ligands. Our data identify an essential function of RNase T2 and RNase 2 upstream of TLR8 and provide insight into TLR8 activation.


Assuntos
Endorribonucleases/metabolismo , Monócitos/imunologia , Neutrófilos/imunologia , RNA Bacteriano/metabolismo , RNA de Protozoário/metabolismo , Receptor 8 Toll-Like/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular , Endorribonucleases/imunologia , Eritrócitos/imunologia , Eritrócitos/parasitologia , Escherichia coli/química , Escherichia coli/imunologia , Edição de Genes/métodos , Humanos , Listeria monocytogenes/química , Listeria monocytogenes/imunologia , Monócitos/microbiologia , Monócitos/parasitologia , Neutrófilos/microbiologia , Neutrófilos/parasitologia , Plasmodium falciparum/química , Plasmodium falciparum/imunologia , Cultura Primária de Células , Estabilidade de RNA , RNA Bacteriano/imunologia , RNA de Protozoário/imunologia , Serratia marcescens/química , Serratia marcescens/imunologia , Staphylococcus aureus/química , Staphylococcus aureus/imunologia , Streptococcus/química , Streptococcus/imunologia , Células THP-1 , Receptor 8 Toll-Like/imunologia
6.
Front Immunol ; 10: 2181, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31572389

RESUMO

Brucella abortus, the causative agent of brucellosis, displays many resources to evade T cell responses conducive to persist inside the host. Our laboratory has previously showed that infection of human monocytes with B. abortus down-modulates the IFN-γ-induced MHC-II expression. Brucella outer membrane lipoproteins are structural components involved in this phenomenon. Moreover, IL-6 is the soluble factor that mediated MHC-II down-regulation. Yet, the MHC-II down-regulation exerted by lipoproteins was less marked than the one observed as consequence of infection. This led us to postulate that there should be other components associated with viable bacteria that may act together with lipoproteins in order to diminish MHC-II. Our group has recently demonstrated that B. abortus RNA (PAMP related to pathogens' viability or vita-PAMP) is involved in MHC-I down-regulation. Therefore, in this study we investigated if B. abortus RNA could be contributing to the down-regulation of MHC-II. This PAMP significantly down-modulated the IFN-γ-induced MHC-II surface expression on THP-1 cells as well as in primary human monocytes and murine bone marrow macrophages. The expression of other molecules up-regulated by IFN-γ (such as co-stimulatory molecules) was stimulated on monocytes treated with B. abortus RNA. This result shows that this PAMP does not alter all IFN-γ-induced molecules globally. We also showed that other bacterial and parasitic RNAs caused MHC-II surface expression down-modulation indicating that this phenomenon is not restricted to B. abortus. Moreover, completely degraded RNA was also able to reproduce the phenomenon. MHC-II down-regulation on monocytes treated with RNA and L-Omp19 (a prototypical lipoprotein of B. abortus) was more pronounced than in monocytes stimulated with both components separately. We also demonstrated that B. abortus RNA along with its lipoproteins decrease MHC-II surface expression predominantly by a mechanism of inhibition of MHC-II expression. Regarding the signaling pathway, we demonstrated that IL-6 is a soluble factor implicated in B. abortus RNA and lipoproteins-triggered MHC-II surface down-regulation. Finally, CD4+ T cells functionality was affected as macrophages treated with these components showed lower antigen presentation capacity. Therefore, B. abortus RNA and lipoproteins are two PAMPs that contribute to MHC-II down-regulation on monocytes/macrophages diminishing CD4+ T cell responses.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Macrófagos/imunologia , Monócitos/imunologia , RNA Bacteriano/imunologia , Animais , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas da Membrana Bacteriana Externa/metabolismo , Brucella abortus/genética , Brucella abortus/imunologia , Brucella abortus/fisiologia , Brucelose/imunologia , Brucelose/microbiologia , Linfócitos T CD4-Positivos/metabolismo , Células Cultivadas , Regulação para Baixo/imunologia , Feminino , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Lipoproteínas/imunologia , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Moléculas com Motivos Associados a Patógenos/imunologia , Moléculas com Motivos Associados a Patógenos/metabolismo , RNA Bacteriano/genética , Células THP-1
7.
mBio ; 10(5)2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31594810

RESUMO

Cellular sensing of bacterial RNA is increasingly recognized as a determinant of host-pathogen interactions. The intracellular pathogen Listeria monocytogenes induces high levels of type I interferons (alpha/beta interferons [IFN-α/ß]) to create a growth-permissive microenvironment during infection. We previously demonstrated that RNAs secreted by L. monocytogenes (comprising the secRNome) are potent inducers of IFN-ß. We determined the composition and diversity of the members of the secRNome and found that they are uniquely enriched for noncoding small RNAs (sRNAs). Testing of individual sRNAs for their ability to induce IFN revealed several sRNAs with this property. We examined ril32, an intracellularly expressed sRNA that is highly conserved for the species L. monocytogenes and that was the most potent inducer of IFN-ß expression of all the sRNAs tested in this study, in more detail. The rli32-induced IFN-ß response is RIG-I (retinoic acid inducible gene I) dependent, and cells primed with rli32 inhibit influenza virus replication. We determined the rli32 motif required for IFN induction. rli32 overproduction promotes intracellular bacterial growth, and a mutant lacking rli32 is restricted for intracellular growth in macrophages. rli32-overproducing bacteria are resistant to H2O2 and exhibit both increased catalase activity and changes in the cell envelope. Comparative transcriptome sequencing (RNA-Seq) analysis indicated that ril32 regulates expression of the lhrC locus, previously shown to be involved in cell envelope stress. Inhibition of IFN-ß signaling by ruxolitinib reduced rli32-dependent intracellular bacterial growth, indicating a link between induction of the interferon system and bacterial physiology. rli32 is, to the best of our knowledge, the first secreted individual bacterial sRNA known to trigger the induction of the type I IFN response.IMPORTANCE Interferons are potent and broadly acting cytokines that stimulate cellular responses to nucleic acids of unusual structures or locations. While protective when induced following viral infections, the induction of interferons is detrimental to the host during L. monocytogenes infection. Here, we identify specific sRNAs, secreted by the bacterium, with the capacity to induce type I IFN. Further analysis of the most potent sRNA, rli32, links the ability to induce RIG-I-dependent induction of the type I IFN response to the intracellular growth properties of the bacterium. Our findings emphasize the significance of released RNA for Listeria infection and shed light on a compartmental strategy used by an intracellular pathogen to modulate host responses to its advantage.


Assuntos
Fatores Imunológicos/metabolismo , Interferon beta/metabolismo , Listeria monocytogenes/imunologia , Listeria monocytogenes/metabolismo , Macrófagos/microbiologia , RNA Bacteriano/metabolismo , Pequeno RNA não Traduzido/metabolismo , Animais , Células Cultivadas , Deleção de Genes , Listeria monocytogenes/genética , Camundongos Endogâmicos C57BL , RNA Bacteriano/genética , RNA Bacteriano/imunologia , Pequeno RNA não Traduzido/genética , Pequeno RNA não Traduzido/imunologia
8.
Front Immunol ; 10: 198, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30846984

RESUMO

Streptococcus pyogenes is a major human pathogen causing a variety of diseases ranging from common pharyngitis to life-threatening soft tissue infections and sepsis. Microbial nucleic acids, especially bacterial RNA, have recently been recognized as a major group of pathogen-associated molecular patterns (PAMPs) involved in the detection of Streptococcus pyogenes via endosomal Toll-like receptors (TLRs) in vitro. However, the individual contribution and cooperation between TLRs as well as cell-type and strain specific differences in dependency on nucleic acid detection during S. pyogenes infection in vitro have not been clarified in detail. Moreover, the role of particularly bacterial RNA for the defense of S. pyogenes infection in vivo remains poorly defined. In this study, we report that in all investigated innate immune cells involved in the resolution of bacterial infections, including murine macrophages, dendritic cells and neutrophils, recognition of S. pyogenes strain ATCC12344 is almost completely dependent on nucleic acid sensing via endosomal TLRs at lower MOIs, whereas at higher MOIs, detection via TLR2 plays an additional, yet redundant role. We further demonstrate that different S. pyogenes strains display a considerable inter-strain variability with respect to their nucleic acid dependent recognition. Moreover, TLR13-dependent recognition of S. pyogenes RNA is largely non-redundant in bone marrow-derived macrophages (BMDMs), but less relevant in neutrophils and bone marrow-derived myeloid dendritic cells (BMDCs) for the induction of an innate immune response in vitro. In vivo, we show that a loss of nucleic acid sensing blunts early recognition of S. pyogenes, leading to a reduced local containment of the bacterial infection with subsequent pronounced systemic inflammation at later time points. Thus, our results argue for a crucial role of nucleic acid sensing via endosomal TLRs in defense of S. pyogenes infection both in vitro and in vivo.


Assuntos
Endossomos/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Infecções Estreptocócicas/imunologia , Infecções Estreptocócicas/metabolismo , Streptococcus pyogenes/fisiologia , Receptores Toll-Like/metabolismo , Biomarcadores , Citocinas/metabolismo , Humanos , Imunidade Celular , Imunidade Inata , Óxido Nítrico/metabolismo , Ácidos Nucleicos/imunologia , RNA Bacteriano/imunologia , Espécies Reativas de Oxigênio/metabolismo , Infecções Estreptocócicas/microbiologia
9.
EMBO Rep ; 20(3)2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30683680

RESUMO

Extracellular vesicles (EVs) have been shown to carry microbial components and function in the host defense against infections. In this study, we demonstrate that Mycobacterium tuberculosis (M.tb) RNA is delivered into macrophage-derived EVs through an M.tb SecA2-dependent pathway and that EVs released from M.tb-infected macrophages stimulate a host RIG-I/MAVS/TBK1/IRF3 RNA sensing pathway, leading to type I interferon production in recipient cells. These EVs also promote, in a RIG-I/MAVS-dependent manner, the maturation of M.tb-containing phagosomes through a noncanonical LC3 pathway, leading to increased bacterial killing. Moreover, treatment of M.tb-infected macrophages or mice with a combination of moxifloxacin and EVs, isolated from M.tb-infected macrophages, significantly lowered bacterial burden relative to either treatment alone. We hypothesize that EVs, which are preferentially removed by macrophages in vivo, can be combined with effective antibiotics as a novel approach to treat drug-resistant TB.


Assuntos
Vesículas Extracelulares/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Imunidade , Viabilidade Microbiana , Mycobacterium tuberculosis/metabolismo , RNA Bacteriano/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Sistemas de Secreção Bacterianos/efeitos dos fármacos , Proteína DEAD-box 58/metabolismo , Vesículas Extracelulares/efeitos dos fármacos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Imunidade/efeitos dos fármacos , Interferon Tipo I/biossíntese , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Viabilidade Microbiana/efeitos dos fármacos , Moxifloxacina/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/crescimento & desenvolvimento , Fator 88 de Diferenciação Mieloide/metabolismo , Fagocitose/efeitos dos fármacos , Fagossomos/efeitos dos fármacos , Fagossomos/metabolismo , Transdução de Sinais/efeitos dos fármacos
10.
Mol Cell ; 73(2): 264-277.e5, 2019 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-30503773

RESUMO

Type ΙΙΙ CRISPR-Cas systems provide robust immunity against foreign RNA and DNA by sequence-specific RNase and target RNA-activated sequence-nonspecific DNase and RNase activities. We report on cryo-EM structures of Thermococcus onnurineus CsmcrRNA binary, CsmcrRNA-target RNA and CsmcrRNA-target RNAanti-tag ternary complexes in the 3.1 Å range. The topological features of the crRNA 5'-repeat tag explains the 5'-ruler mechanism for defining target cleavage sites, with accessibility of positions -2 to -5 within the 5'-repeat serving as sensors for avoidance of autoimmunity. The Csm3 thumb elements introduce periodic kinks in the crRNA-target RNA duplex, facilitating cleavage of the target RNA with 6-nt periodicity. Key Glu residues within a Csm1 loop segment of CsmcrRNA adopt a proposed autoinhibitory conformation suggestive of DNase activity regulation. These structural findings, complemented by mutational studies of key intermolecular contacts, provide insights into CsmcrRNA complex assembly, mechanisms underlying RNA targeting and site-specific periodic cleavage, regulation of DNase cleavage activity, and autoimmunity suppression.


Assuntos
Autoimunidade , Proteínas de Bactérias/metabolismo , Proteínas Associadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Desoxirribonucleases/metabolismo , Estabilidade de RNA , RNA Bacteriano/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/ultraestrutura , Proteínas Associadas a CRISPR/genética , Proteínas Associadas a CRISPR/imunologia , Proteínas Associadas a CRISPR/ultraestrutura , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/imunologia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/imunologia , Microscopia Crioeletrônica , Desoxirribonucleases/genética , Desoxirribonucleases/imunologia , Desoxirribonucleases/ultraestrutura , Escherichia coli/enzimologia , Escherichia coli/genética , Escherichia coli/imunologia , Regulação Bacteriana da Expressão Gênica , Modelos Moleculares , Complexos Multiproteicos , Mutação , Conformação de Ácido Nucleico , Conformação Proteica , RNA Bacteriano/genética , RNA Bacteriano/imunologia , RNA Bacteriano/ultraestrutura , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Proteínas de Ligação a RNA/ultraestrutura , Relação Estrutura-Atividade , Thermococcus/enzimologia , Thermococcus/genética , Thermococcus/imunologia
11.
Mol Cell ; 73(2): 278-290.e4, 2019 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-30503774

RESUMO

Adaptive immune systems must accurately distinguish between self and non-self in order to defend against invading pathogens while avoiding autoimmunity. Type III CRISPR-Cas systems employ guide RNA to recognize complementary RNA targets, which triggers the degradation of both the invader's transcripts and their template DNA. These systems can broadly eliminate foreign targets with multiple mutations but circumvent damage to the host genome. To explore the molecular basis for these features, we use single-molecule fluorescence microscopy to study the interaction between a type III-A ribonucleoprotein complex and various RNA substrates. We find that Cas10-the DNase effector of the complex-displays rapid conformational fluctuations on foreign RNA targets, but is locked in a static configuration on self RNA. Target mutations differentially modulate Cas10 dynamics and tune the CRISPR interference activity in vivo. These findings highlight the central role of the internal dynamics of CRISPR-Cas complexes in self versus non-self discrimination and target specificity.


Assuntos
Autoimunidade , Proteínas de Bactérias/imunologia , Proteínas Associadas a CRISPR/imunologia , Sistemas CRISPR-Cas/imunologia , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/imunologia , RNA Bacteriano/imunologia , Tolerância a Antígenos Próprios , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas Associadas a CRISPR/genética , Proteínas Associadas a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Escherichia coli/enzimologia , Escherichia coli/genética , Escherichia coli/imunologia , Cinética , Microscopia de Fluorescência , Mutação , Conformação de Ácido Nucleico , Conformação Proteica , RNA Bacteriano/química , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , Transdução de Sinais , Imagem Individual de Molécula/métodos , Staphylococcus aureus/enzimologia , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia , Staphylococcus epidermidis/enzimologia , Staphylococcus epidermidis/genética , Staphylococcus epidermidis/imunologia , Relação Estrutura-Atividade
12.
J Immunol ; 200(9): 3244-3258, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29610140

RESUMO

Upon recognition of a microbial pathogen, the innate and adaptive immune systems are linked to generate a cell-mediated immune response against the foreign invader. The culture filtrate of Mycobacterium tuberculosis contains ligands, such as M. tuberculosis tRNA, that activate the innate immune response and secreted Ags recognized by T cells to drive adaptive immune responses. In this study, bioinformatics analysis of gene-expression profiles derived from human PBMCs treated with distinct microbial ligands identified a mycobacterial tRNA-induced innate immune network resulting in the robust production of IL-12p70, a cytokine required to instruct an adaptive Th1 response for host defense against intracellular bacteria. As validated by functional studies, this pathway contained a feed-forward loop, whereby the early production of IL-18, type I IFNs, and IL-12p70 primed NK cells to respond to IL-18 and produce IFN-γ, enhancing further production of IL-12p70. Mechanistically, tRNA activates TLR3 and TLR8, and this synergistic induction of IL-12p70 was recapitulated by the addition of a specific TLR8 agonist with a TLR3 ligand to PBMCs. These data indicate that M. tuberculosis tRNA activates a gene network involving the integration of multiple innate signals, including types I and II IFNs, as well as distinct cell types to induce IL-12p70.


Assuntos
Interleucina-12/imunologia , Mycobacterium tuberculosis/imunologia , RNA Bacteriano/imunologia , RNA de Transferência/imunologia , Tuberculose/imunologia , Diferenciação Celular/imunologia , Redes Reguladoras de Genes/imunologia , Humanos , Imunidade Celular/imunologia , Imunidade Inata/imunologia , Interleucina-12/biossíntese , Ativação Linfocitária/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Células Th1/imunologia
13.
Immunity ; 48(3): 584-598.e5, 2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29548673

RESUMO

Live vaccines historically afford superior protection, yet the cellular and molecular mechanisms mediating protective immunity remain unclear. Here we found that vaccination of mice with live, but not dead, Gram-negative bacteria heightened follicular T helper cell (Tfh) differentiation, germinal center formation, and protective antibody production through the signaling adaptor TRIF. Complementing the dead vaccine with an innate signature of bacterial viability, bacterial RNA, recapitulated these responses. The interferon (IFN) and inflammasome pathways downstream of TRIF orchestrated Tfh responses extrinsically to B cells and classical dendritic cells. Instead, CX3CR1+CCR2- monocytes instructed Tfh differentiation through interleukin-1ß (IL-1ß), a tightly regulated cytokine secreted upon TRIF-dependent IFN licensing of the inflammasome. Hierarchical production of IFN-ß and IL-1ß dictated Tfh differentiation and elicited the augmented humoral responses characteristic of live vaccines. These findings identify bacterial RNA, an innate signature of microbial viability, as a trigger for Tfh differentiation and suggest new approaches toward vaccine formulations for coordinating augmented Tfh and B cell responses.


Assuntos
Formação de Anticorpos/imunologia , Linfócitos B/imunologia , Ativação Linfocitária/imunologia , Viabilidade Microbiana/imunologia , RNA Bacteriano/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Anticorpos Neutralizantes/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Linfócitos B/metabolismo , Vacinas Bacterianas/imunologia , Biomarcadores , Diferenciação Celular/imunologia , Citocinas/metabolismo , Centro Germinativo , Interações Hospedeiro-Patógeno/imunologia , Imunidade Celular , Imunidade Inata , Inflamassomos/metabolismo , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo , Transdução de Sinais , Linfócitos T Auxiliares-Indutores/metabolismo
14.
Front Immunol ; 9: 27, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29410667

RESUMO

Lactic acid bacteria (LAB) are one of the major commensal species in the small intestine and known for contributing to maintenance of protective immunity and immune homeostasis. However, currently there has been no evidence regarding the cellular mechanisms involved in the probiotic effects of LAB on human immune cells. Here, we demonstrated that LAB double-stranded RNA (dsRNA) triggered interferon-ß (IFN-ß) production by human dendritic cells (DCs), which activated IFN-γ-producing T cells. Interleukin-12 (IL-12) secretion from human DCs in response to LAB was abrogated by depletion of bacterial dsRNA, and was attenuated by neutralizing IFN-ß, indicating LAB dsRNA primarily activated the IFN-ß/IL-12 pathway. Moreover, the induction of IL-12 secretion from DCs by LAB was abolished by the inhibition of endosomal acidification, confirming the critical role of the endosomal digestion of LAB. In a coculture of human naïve CD4+ T cells and BDCA1+ DCs, DCs stimulated with LAB containing dsRNA induced IFN-γ-producing T cells. These results indicate that human DCs activated by LAB enhance Th1 immunity depending on IFN-ß secretion in response to bacterial dsRNA.


Assuntos
Células Dendríticas/imunologia , Interferon beta/imunologia , Lactobacillales/imunologia , RNA de Cadeia Dupla/imunologia , Células Th1/imunologia , Antígenos CD1/metabolismo , Células Cultivadas , Glicoproteínas/metabolismo , Humanos , Interferon beta/biossíntese , Interferon gama/biossíntese , Interferon gama/imunologia , Subunidade p35 da Interleucina-12/metabolismo , Lactobacillales/genética , RNA Bacteriano/imunologia , Células Th2/metabolismo
15.
Microb Pathog ; 114: 219-224, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29180292

RESUMO

The molecular basis of intraocular tuberculosis (TB) is not well understood. In this study, we investigated the role of two constituents of viable Mycobacterium tuberculosis - Early Secreted Antigenic Target-6 (ESAT-6), and mycobacterial RNA- in inflammasome activation in the retinal pigment epithelium (RPE), a key site of inflammation in intraocular TB. We found that ESAT-6 induced caspase-1 activation and inflammasome priming in mouse RPE cells, substantially more in wild-type than in Tlr2/3/4/7/9-/-, Myd88-/- or Nlrp3-/- RPE cells. Sub-retinal ESAT-6 injection resulted in greater RPE degeneration in wild-type than in Nlrp3-/- mice. In human ocular TB tissue sections, NLRP3 staining was noted in retina as well as RPE. Mycobacterial RNA, specifically its double stranded component, also induced caspase-1 activation, and the double stranded RNA was immunolocalized to human ocular TB sections. Our observations suggest that inflammasome activation in RPE by viable M. tuberculosis could potentially contribute to human intraocular TB.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas de Bactérias/imunologia , Inflamassomos/imunologia , Mycobacterium tuberculosis/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , RNA Bacteriano/imunologia , RNA de Cadeia Dupla/imunologia , Tuberculose Ocular/imunologia , Animais , Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Caspase 1/genética , Caspase 1/imunologia , Interações Hospedeiro-Parasita , Humanos , Inflamassomos/genética , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium tuberculosis/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , RNA Bacteriano/genética , RNA de Cadeia Dupla/genética , Epitélio Pigmentado da Retina/imunologia , Epitélio Pigmentado da Retina/microbiologia , Tuberculose Ocular/genética , Tuberculose Ocular/microbiologia
16.
J Allergy Clin Immunol ; 141(1): 94-103.e15, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28479329

RESUMO

BACKGROUND: Asthma pathophysiology and treatment responsiveness are predicted by inflammatory phenotype. However, the relationship between airway microbiology and asthma phenotype is poorly understood. OBJECTIVE: We aimed to characterize the airway microbiota in patients with symptomatic stable asthma and relate composition to airway inflammatory phenotype and other phenotypic characteristics. METHODS: The microbial composition of induced sputum specimens collected from adult patients screened for a multicenter randomized controlled trial was determined by using 16S rRNA gene sequencing. Inflammatory phenotypes were defined by sputum neutrophil and eosinophil cell proportions. Microbiota were defined by using α- and ß-diversity measures, and interphenotype differences were identified by using similarity of percentages, network analysis, and taxon fold change. Phenotypic predictors of airway microbiology were identified by using multivariate linear regression. RESULTS: Microbiota composition was determined in 167 participants and classified as eosinophilic (n = 84), neutrophilic (n = 14), paucigranulocytic (n = 60), or mixed neutrophilic-eosinophilic (n = 9) asthma phenotypes. Airway microbiology was significantly less diverse (P = .022) and more dissimilar (P = .005) in neutrophilic compared with eosinophilic participants. Sputum neutrophil proportions, but not eosinophil proportions, correlated significantly with these diversity measures (α-diversity: Spearman r = -0.374, P < .001; ß-diversity: r = 0.238, P = .002). Interphenotype differences were characterized by a greater frequency of pathogenic taxa at high relative abundance and reduced Streptococcus, Gemella, and Porphyromonas taxa relative abundance in patients with neutrophilic asthma. Multivariate regression confirmed that sputum neutrophil proportion was the strongest predictor of microbiota composition. CONCLUSIONS: Neutrophilic asthma is associated with airway microbiology that is significantly different from that seen in patients with other inflammatory phenotypes, particularly eosinophilic asthma. Differences in microbiota composition might influence the response to antimicrobial and steroid therapies and the risk of lung infection.


Assuntos
Asma , Bactérias , Microbiota , RNA Bacteriano , RNA Ribossômico 16S , Adulto , Idoso , Asma/imunologia , Asma/microbiologia , Bactérias/classificação , Bactérias/genética , Bactérias/imunologia , Feminino , Humanos , Masculino , Microbiota/genética , Microbiota/imunologia , Pessoa de Meia-Idade , Neutrófilos/imunologia , RNA Bacteriano/genética , RNA Bacteriano/imunologia , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/imunologia , Índice de Gravidade de Doença
17.
PLoS Pathog ; 13(8): e1006527, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28767704

RESUMO

Despite eliciting a potent CD8+ T cell response, Brucella abortus is able to persist and establish a chronic infection inside its host. We have previously reported that the infection of human monocytes/macrophages with B. abortus inhibits the IFN-γ-induced MHC-I cell surface expression down-modulating cytotoxic CD8+ T cell responses. MHC-I down-modulation depends on bacterial viability and results from the capacity of B. abortus to retain the MHC-I molecules within the Golgi apparatus. Furthermore, we recently demonstrated that epidermal growth factor receptor (EGFR) pathway is involved in this phenomenon and that this is an early event during infection. However, the components and mechanisms whereby B. abortus is able to down-modulate MHC-I remained to be elucidated. In this study we demonstrated that the down-modulation of MHC-I expression is not mediated by well-known Brucella virulence factors but instead by B. abortus RNA, a PAMP associated to viability (vita-PAMP). Surprisingly, completely degraded RNA was also able to inhibit MHC-I expression to the same extent as intact RNA. Accordingly, B. abortus RNA and its degradation products were able to mimic the MHC-I intracellular retention within the Golgi apparatus observed upon infection. We further demonstrated that TLR8, a single-stranded RNA and RNA degradation products sensor, was involved in MHC-I inhibition. On the other hand, neutralization of the EGFR reversed the MHC-I inhibition, suggesting a connection between the TLR8 and EGFR pathways. Finally, B. abortus RNA-treated macrophages display diminished capacity of antigen presentation to CD8+ T cells. Overall, our results indicate that the vita-PAMP RNA as well as its degradation products constitute novel virulence factors whereby B. abortus, by a TLR8-dependent mechanism and through the EGFR pathway, inhibits the IFN-γ-induced MHC-I surface expression on human monocytes/macrophages. Thus, bacteria can hide within infected cells and avoid the immunological surveillance of cytotoxic CD8+ T cells.


Assuntos
Brucelose/imunologia , Receptores ErbB/imunologia , Evasão da Resposta Imune/imunologia , Monócitos/imunologia , RNA Bacteriano/imunologia , Receptor 8 Toll-Like/imunologia , Animais , Brucella abortus/imunologia , Apresentação Cruzada/imunologia , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/biossíntese , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Monócitos/microbiologia , Transdução de Sinais/imunologia
18.
Artigo em Inglês | MEDLINE | ID: mdl-28730145

RESUMO

Polymorphonuclear neutrophils (PMN) are the first cellular line of antibacterial host defense. They sense pathogens through recognition of pathogen-associated molecular patterns (PAMPs) by innate pattern recognition receptors, such as Toll-like receptors (TLR). The aim of this study was to investigate whether PMN sense bacterial viability and explore which viability factor could be involved in this phenomenon. For this purpose, different functions were evaluated in isolated human PMN using live Escherichia coli (Ec) and heat-killed Ec (HK-Ec). We found that bacterial viability was indispensable to induce PMN activation, as measured by forward-scatter (FSC) increase, CD11b surface expression, chemotaxis, reactive oxygen species (ROS) generation and neutrophil extracellular trap (NET) formation. As uncapped non-polyadenylated prokaryotic mRNA has been recognized as a PAMP associated to bacterial viability by macrophages and dendritic cells, total prokaryotic RNA (pRNA) from live Ec was purified and used as a stimulus for PMN. pRNA triggered similar responses to those observed with live bacteria. No RNA could be isolated from HK-Ec, explaining the lack of effect of dead bacteria. Moreover, the supernatant of dead bacteria was able to induce PMN activation, and this was associated with the presence of pRNA in this supernatant, which is released in the killing process. The induction of bactericidal functions (ROS and NETosis) by pRNA were abolished when the supernatant of dead bacteria or isolated pRNA were treated with RNAse. Moreover, endocytosis was necessary for pRNA-induced ROS generation and NETosis, and priming was required for the induction of pRNA-induced ROS in whole blood. However, responses related to movement and degranulation (FSC increase, CD11b up-regulation, and chemotaxis) were still triggered when pRNA was digested with RNase, and were not dependent on pRNA endocytosis or PMN priming. In conclusion, our results indicate that PMN sense live bacteria through recognition of pRNA, and this sensing triggers potent bactericidal mechanisms.


Assuntos
Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Neutrófilos/imunologia , RNA Bacteriano/imunologia , Antígeno CD11b/genética , Antígeno CD11b/imunologia , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Infecções por Escherichia coli/genética , Infecções por Escherichia coli/microbiologia , Humanos , Viabilidade Microbiana , Ativação de Neutrófilo , Neutrófilos/microbiologia , RNA Bacteriano/genética
19.
Biosci Biotechnol Biochem ; 81(6): 1099-1105, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28406067

RESUMO

Enterococcus faecalis is a resident lactic acid bacterium in the human intestine. Its immunostimulatory action was reported to be enhanced by heat sterilization. To investigate its beneficial actions, we evaluated the ability of 10 E. faecalis strains to induce interleukin-12 (IL-12) production in a mouse macrophage cell line, J774.1 and found that the strain, E. faecalis IC-1, had a potent IL-12-inducing ability. Furthermore, we investigated the underlying mechanism by treating IC-1 cells with RNase or lysozyme. Its activity almost disappeared and an antagonist of Toll-like receptor (TLR) 7 inhibited this activity. Moreover, lysozyme-treated IC-1 bacteria were not phagocytized by J774.1 cells, and did not induce IL-12 production. Based on our results, we propose that macrophages recognize the cell wall components of IC-1, leading to phagocytosis. The IC-1 RNA is then recognized by TLR7, which induces the production of IL-12.


Assuntos
Parede Celular/imunologia , Enterococcus faecalis/imunologia , Interleucina-12/imunologia , Macrófagos/imunologia , RNA Bacteriano/imunologia , Animais , Linhagem Celular , Parede Celular/química , Parede Celular/efeitos dos fármacos , Técnicas de Cocultura , Enterococcus faecalis/química , Enterococcus faecalis/efeitos dos fármacos , Expressão Gênica , Interleucina-12/biossíntese , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Muramidase/química , Muramidase/farmacologia , Oligonucleotídeos/farmacologia , Fagocitose/efeitos dos fármacos , RNA Bacteriano/química , Ribonucleases/química , Ribonucleases/farmacologia , Receptor 7 Toll-Like/antagonistas & inibidores , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/imunologia
20.
J Exp Med ; 214(5): 1297-1311, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28400473

RESUMO

The spores of pathogenic bacteria are involved in host entry and the initial encounter with the host immune system. How bacterial spores interact with host immunity, however, remains poorly understood. Here, we show that the spores of Bacillus anthracis (BA), the etiologic agent of anthrax, possess an intrinsic ability to induce host immune responses. This immunostimulatory activity is attributable to high amounts of RNA present in the spore surface layer. RNA-sensing TLRs, TLR7, and TLR13 in mice and their human counterparts, are responsible for detecting and triggering the host cell response to BA spores, whereas TLR2 mediates the sensing of vegetative BA. BA spores, but not vegetative BA, induce type I IFN (IFN-I) production. Although TLR signaling in itself affords protection against BA, spore RNA-induced IFN-I signaling is disruptive to BA clearance. Our study suggests a role for bacterial spore-associated RNA in microbial pathogenesis and illustrates a little known aspect of interactions between the host and spore-forming bacteria.


Assuntos
RNA Bacteriano/imunologia , Esporos Bacterianos/imunologia , Receptores Toll-Like/fisiologia , Animais , Bacillus anthracis/imunologia , Regulação da Expressão Gênica/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Camundongos , Esporos Bacterianos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...