Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
Front Immunol ; 13: 980400, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36389822

RESUMO

Sjögren's syndrome (SS) is a chronic systemic autoimmune disease that affects the salivary and lacrimal glands, as well as other organ systems like the lungs, kidneys and nervous system. SS can occur alone or in combination with another autoimmune disease, such as systemic lupus erythematosus (SLE) or rheumatoid arthritis. The etiology of SS is unknown but recent studies have revealed the implication of the activation of innate immune receptors, including Toll-like receptors (TLRs), mainly through the detection of endogenous nucleic acids, in the pathogenesis of systemic autoimmune diseases. Studies on SS mouse models suggest that TLRs and especially TLR7 that detects single-stranded RNA of microbial or endogenous origin can drive the development of SS and findings in SS patients corroborate those in mouse models. In this review, we will give an overview of the function and signaling of nucleic acid-sensing TLRs, the interplay of TLR7 with TLR8 and TLR9 in the context of autoimmunity, summarize the evidence for the critical role of TLR7 in the pathogenesis of SS and present a possible connection between SARS-CoV-2 and SS.


Assuntos
COVID-19 , Ácidos Nucleicos , Síndrome de Sjogren , Camundongos , Animais , Receptor 7 Toll-Like/genética , SARS-CoV-2 , Receptores Toll-Like
2.
Nat Immunol ; 23(3): 458-468, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35210623

RESUMO

Alveolar macrophages (AMs) are lung tissue-resident macrophages that can be expanded in culture, but it is unknown to what extent culture affects their in vivo identity. Here we show that mouse long-term ex vivo expanded AMs (exAMs) maintained a core AM gene expression program, but showed culture adaptations related to adhesion, metabolism and proliferation. Upon transplantation into the lung, exAMs reacquired full transcriptional and epigenetic AM identity, even after several months in culture and could self-maintain long-term in the alveolar niche. Changes in open chromatin regions observed in culture were fully reversible in transplanted exAMs and resulted in a gene expression profile indistinguishable from resident AMs. Our results indicate that long-term proliferation of AMs in culture did not compromise cellular identity in vivo. The robustness of exAM identity provides new opportunities for mechanistic analysis and highlights the therapeutic potential of exAMs.


Assuntos
Pulmão , Macrófagos Alveolares , Animais , Cromatina/metabolismo , Epigênese Genética , Epigenômica , Pulmão/metabolismo , Macrófagos Alveolares/metabolismo , Camundongos
3.
Front Immunol ; 12: 676010, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34108972

RESUMO

Sjögren's syndrome (SS) is a chronic systemic autoimmune disease that affects predominately salivary and lacrimal glands. SS can occur alone or in combination with another autoimmune disease like systemic lupus erythematosus (SLE). Here we report that TLR7 signaling drives the development of SS since TLR8-deficient (TLR8ko) mice that develop lupus due to increased TLR7 signaling by dendritic cells, also develop an age-dependent secondary pathology similar to associated SS. The SS phenotype in TLR8ko mice is manifested by sialadenitis, increased anti-SSA and anti-SSB autoantibody production, immune complex deposition and increased cytokine production in salivary glands, as well as lung inflammation. Moreover, ectopic lymphoid structures characterized by B/T aggregates, formation of high endothelial venules and the presence of dendritic cells are formed in the salivary glands of TLR8ko mice. Interestingly, all these phenotypes are abrogated in double TLR7/8-deficient mice, suggesting that the SS phenotype in TLR8-deficient mice is TLR7-dependent. In addition, evaluation of TLR7 and inflammatory markers in the salivary glands of primary SS patients revealed significantly increased TLR7 expression levels compared to healthy individuals, that were positively correlated to TNF, LT-α, CXCL13 and CXCR5 expression. These findings establish an important role of TLR7 signaling for local and systemic SS disease manifestations, and inhibition of such will likely have therapeutic value.


Assuntos
Síndrome de Sjogren/etiologia , Receptor 7 Toll-Like/fisiologia , Adulto , Idoso , Animais , Quimiocinas/genética , Citocinas/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Pneumonia/etiologia , Transdução de Sinais/fisiologia , Síndrome de Sjogren/imunologia
4.
Front Immunol ; 10: 2015, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31552019

RESUMO

Systemic lupus erythematosus (SLE) patients have increased prevalence of metabolic syndrome but the underlying mechanisms are unknown. Toll-like receptor 7 (TLR7) that detects single stranded-RNA plays a key role in antimicrobial host defense and also contributes to the initiation and progression of SLE both in mice and humans. Here, we report the implication of TLR7 signaling in high fat diet (HFD)-induced metabolic syndrome and exacerbation of lupus autoimmunity in TLR8-deficient (TLR8ko) mice, which develop spontaneous lupus-like disease due to increased TLR7 signaling by dendritic cells (DCs). The aggravated SLE pathogenesis in HFD-fed TLR8ko mice was characterized by increased overall immune activation, anti-DNA autoantibody production, and IgG/IgM glomerular deposition that were coupled with increased kidney histopathology. Moreover, upon HFD TLR8ko mice developed metabolic abnormalities, including liver inflammation. In contrast, upon HFD TLR7/8ko mice did not develop SLE and both TLR7ko and TLR7/8ko mice were fully protected from metabolic abnormalities, including body weight gain, insulin resistance, and liver inflammation. Interestingly, HFD led to an increase of TLR7 expression in WT mice, that was coupled with increased TNF production by DCs, and this phenotype was more profound in TLR8ko mice. Our study uncovers the implication of TLR7 signaling in the interconnection of SLE and metabolic abnormalities, indicating that TLR7 might be a novel approach as a tailored therapy in SLE and metabolic diseases.


Assuntos
Lúpus Eritematoso Sistêmico/imunologia , Obesidade/imunologia , Transdução de Sinais/imunologia , Receptor 7 Toll-Like/imunologia , Animais , Anticorpos Antinucleares/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Dieta Hiperlipídica/efeitos adversos , Humanos , Resistência à Insulina/imunologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/metabolismo , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/etiologia , Obesidade/metabolismo , Transdução de Sinais/genética , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/genética , Receptor 8 Toll-Like/imunologia , Receptor 8 Toll-Like/metabolismo , Aumento de Peso/imunologia
5.
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
6.
J Immunol ; 197(11): 4425-4435, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27798161

RESUMO

West Nile virus (WNV) is a neurotropic ssRNA flavivirus that can cause encephalitis, meningitis, and death in humans and mice. Human TLR7 and TLR8 and mouse TLR7 recognize viral ssRNA motifs and induce antiviral immunity. However, the role of mouse TLR8 in antiviral immunity is poorly understood. In this article, we report that TLR8-deficient (Tlr8-/-) mice were resistant to WNV infection compared with wild-type controls. Efficient WNV clearance and moderate susceptibility to WNV-mediated neuronal death in Tlr8-/- mice were attributed to overexpression of Tlr7 and IFN-stimulated gene-56 expression, whereas reduced expression of the proapoptotic gene coding Bcl2-associated X protein was observed. Interestingly, suppressor of cytokine signaling (SOCS)-1 directly associated with TLR8, but not with TLR7, indicating a novel role for TLR8 regulation of SOCS-1 function, whereas selective small interfering RNA knockdown of Socs-1 resulted in induced IFN-stimulated gene-56 and Tlr7 expression following WNV infection. Collectively, we report that TLR8 coupling with SOCS-1 inhibits TLR7-mediated antiviral immunity during WNV infection in mice.


Assuntos
Proteína 1 Supressora da Sinalização de Citocina/imunologia , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/imunologia , Febre do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/imunologia , Animais , Camundongos , Camundongos Knockout , Proteína 1 Supressora da Sinalização de Citocina/genética , Receptor 7 Toll-Like/genética , Receptor 8 Toll-Like/genética , Febre do Nilo Ocidental/genética
7.
Eur J Immunol ; 46(12): 2801-2811, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27600904

RESUMO

The transcriptional repressor growth factor independence 1 (Gfi1) is important in myeloid and lymphoid differentiation. In the current study we evaluated the involvement of Gfi1 in systemic lupus erythematosus (SLE). We found that Genista mice, which carry a hypomorphic mutation in the gfi1 gene or Gfi1-deficient (Gfi1-/- ) mice develop signs of spontaneous lupus autoimmunity, including increased serum levels of IgM and IgG2a, autoantibodies against RNA and DNA, glomerular immunodeposits and increased frequencies of plasmablasts, germinal center (GC) B cells and age-associated B cells (ABCs). On the contrary, Genista mice deprived of TLR7 did not show any of these phenotypes, suggesting that the observed lupus autoimmunity in Genista mice is TLR7-dependent. Moreover, Genista mice showed an increased activation of dendritic cells (DCs), B and T cells that was dependent on TLR7 for DCs and B cells, but not for T cells. Upon TLR7 or TLR4 stimulation Genista DCs produced increased amounts of TNF, IL-6 and IFN-ß and showed increased NF-κB phosphorylation and IRF7 nuclear translocation, suggesting that Gfi1 controls the NF-κB and type I IFN signaling pathway downstream of TLRs. Our data reveal that Gfi1 plays a critical role in the prevention of spontaneous lupus autoimmunity by negatively regulating TLR7 signaling.


Assuntos
Células da Medula Óssea/imunologia , Proteínas de Ligação a DNA/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Fatores de Transcrição/metabolismo , Animais , Autoimunidade , Células Cultivadas , Proteínas de Ligação a DNA/genética , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais , Receptor 7 Toll-Like/genética , Receptor 8 Toll-Like/genética , Fatores de Transcrição/genética
8.
PLoS One ; 10(3): e0119727, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25756897

RESUMO

Innate immune recognition of the major human-specific Gram-positive pathogen Streptococcus pyogenes is not understood. Here we show that mice employ Toll-like receptor (TLR) 2- and TLR13-mediated recognition of S. pyogenes. These TLR pathways are non-redundant in the in vivo context of animal infection, but are largely redundant in vitro, as only inactivation of both of them abolishes inflammatory cytokine production by macrophages and dendritic cells infected with S. pyogenes. Mechanistically, S. pyogenes is initially recognized in a phagocytosis-independent manner by TLR2 and subsequently by TLR13 upon internalization. We show that the TLR13 response is specifically triggered by S. pyogenes rRNA and that Tlr13-/- cells respond to S. pyogenes infection solely by engagement of TLR2. TLR13 is absent from humans and, remarkably, we find no equivalent route for S. pyogenes RNA recognition in human macrophages. Phylogenetic analysis reveals that TLR13 occurs in all kingdoms but only in few mammals, including mice and rats, which are naturally resistant against S. pyogenes. Our study establishes that the dissimilar expression of TLR13 in mice and humans has functional consequences for recognition of S. pyogenes in these organisms.


Assuntos
Streptococcus pyogenes/imunologia , Receptor 2 Toll-Like/metabolismo , Receptores Toll-Like/metabolismo , Animais , Células HEK293 , Humanos , Imunidade Inata , Proteínas de Membrana Transportadoras/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Filogenia , RNA Bacteriano/imunologia , Receptores Toll-Like/genética
9.
Infect Immun ; 82(12): 5013-22, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25225249

RESUMO

Murine Toll-like receptor 13 (TLR13), an endosomal receptor that is not present in humans, is activated by an unmethylated motif present in the large ribosomal subunit of bacterial RNA (23S rRNA). Little is known, however, of the impact of TLR13 on antibacterial host defenses. Here we examined the role of this receptor in the context of infection induced by the model pathogen group B streptococcus (GBS). To this end, we used bacterial strains masked from TLR13 recognition by virtue of constitutive expression of the ErmC methyltransferase, which results in dimethylation of the 23S rRNA motif at a critical adenine residue. We found that TLR13-mediated rRNA recognition was required for optimal induction of tumor necrosis factor alpha and nitrous oxide in dendritic cell and macrophage cultures stimulated with heat-killed bacteria or purified bacterial RNA. However, TLR13-dependent recognition was redundant when live bacteria were used as a stimulus. Moreover, masking bacterial rRNA from TLR13 recognition did not increase the ability of GBS to avoid host defenses and replicate in vivo. In contrast, increased susceptibility to infection was observed under conditions in which signaling by all endosomal TLRs was abolished, i.e., in mice with a loss-of-function mutation in the chaperone protein UNC93B1. Our data lend support to the conclusion that TLR13 participates in GBS recognition, although blockade of the function of this receptor can be compensated for by other endosomal TLRs. Lack of selective pressure by bacterial infections might explain the evolutionary loss of TLR13 in humans. However, further studies using different bacterial species are needed to prove this hypothesis.


Assuntos
Imunidade Inata , Streptococcus agalactiae/imunologia , Receptores Toll-Like/imunologia , Animais , Células Cultivadas , Análise por Conglomerados , DNA Bacteriano/química , DNA Bacteriano/genética , DNA Ribossômico/química , DNA Ribossômico/genética , Células Dendríticas , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Filogenia , RNA Ribossômico 23S/imunologia , Análise de Sequência de DNA
10.
Methods Mol Biol ; 1197: 209-25, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25172283

RESUMO

Innate immune recognition of microbial components is critical for the onset of an appropriate immune response against invading pathogens, in particular by dendritic cells. Toll-like receptors (TLRs) are key in the detection of a variety of microbial stimuli.Here we focus on the methodology used to evaluate the role of TLRs in the process of dendritic cell response to bacterial intracellular infections, using bone marrow-derived dendritic cells (BMDCs) as a model system. This protocol describes how to access the level of activation of BMDCs using standard immunology and biochemistry approaches along with examination of infected cells by immunofluorescence microscopy.


Assuntos
Infecções Bacterianas/metabolismo , Células Dendríticas/metabolismo , Receptores Toll-Like/metabolismo , Animais , Células da Medula Óssea/citologia , Camundongos , Microscopia de Fluorescência , Transdução de Sinais/fisiologia
11.
Proc Natl Acad Sci U S A ; 111(4): 1497-502, 2014 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-24474776

RESUMO

Systemic lupus erythematosus (SLE) is a complex autoimmune disease with diverse clinical presentations characterized by the presence of autoantibodies to nuclear components. Toll-like receptor (TLR)7, TLR8, and TLR9 sense microbial or endogenous nucleic acids and are implicated in the development of SLE. In mice TLR7-deficiency ameliorates SLE, but TLR8- or TLR9-deficiency exacerbates the disease because of increased TLR7 response. Thus, both TLR8 and TLR9 control TLR7 function, but whether TLR8 and TLR9 act in parallel or in series in the same or different cell types in controlling TLR7-mediated lupus remains unknown. Here, we reveal that double TLR8/9-deficient (TLR8/9(-/-)) mice on the C57BL/6 background showed increased abnormalities characteristic of SLE, including splenomegaly, autoantibody production, frequencies of marginal zone and B1 B cells, and renal pathology compared with single TLR8(-/-) or TLR9(-/-) mice. On the cellular level, TLR8(-/-) and TLR8/9(-/-) dendritic cells were hyperesponsive to TLR7 ligand R848, but TLR9(-/-) cells responded normally. Moreover, B cells from TLR9(-/-) and TLR8/9(-/-) mice were hyperesponsive to R848, but TLR8(-/-) B cells were not. These results reveal that TLR8 and TLR9 have an additive effect on controlling TLR7 function and TLR7-mediated lupus; however, they act on different cell types. TLR8 controls TLR7 function on dendritic cells, and TLR9 restrains TLR7 response on B cells.


Assuntos
Autoimunidade/fisiologia , Linfócitos B/imunologia , Células Dendríticas/imunologia , Glicoproteínas de Membrana/fisiologia , Receptor 7 Toll-Like/fisiologia , Receptor 8 Toll-Like/fisiologia , Receptor Toll-Like 9/fisiologia , Animais , Citometria de Fluxo , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Receptor 7 Toll-Like/genética , Receptor 8 Toll-Like/genética , Receptor Toll-Like 9/genética
12.
Artigo em Inglês | MEDLINE | ID: mdl-23847770

RESUMO

Several bacterial pathogens have TIR domain-containing proteins that contribute to their pathogenesis. We identified a second TIR-containing protein in Brucella spp. that we have designated BtpB. We show it is a potent inhibitor of TLR signaling, probably via MyD88. BtpB is a novel Brucella effector that is translocated into host cells and interferes with activation of dendritic cells. In vivo mouse studies revealed that BtpB is contributing to virulence and control of local inflammatory responses with relevance in the establishment of chronic brucellosis. Together, our results show that BtpB is a novel Brucella effector that plays a major role in the modulation of host innate immune response during infection.


Assuntos
Proteínas de Bactérias/metabolismo , Brucella/imunologia , Brucella/patogenicidade , Evasão da Resposta Imune , Fatores de Virulência/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Bactérias/imunologia , Brucelose/imunologia , Brucelose/microbiologia , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Alinhamento de Sequência , Transdução de Sinais , Análise de Sobrevida , Receptores Toll-Like/imunologia , Fatores de Virulência/imunologia
13.
Nature ; 491(7426): 774-8, 2012 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-23103862

RESUMO

The mammalian host has developed a long-standing symbiotic relationship with a considerable number of microbial species. These include the microbiota on environmental surfaces, such as the respiratory and gastrointestinal tracts, and also endogenous retroviruses (ERVs), comprising a substantial fraction of the mammalian genome. The long-term consequences for the host of interactions with these microbial species can range from mutualism to parasitism and are not always completely understood. The potential effect of one microbial symbiont on another is even less clear. Here we study the control of ERVs in the commonly used C57BL/6 (B6) mouse strain, which lacks endogenous murine leukaemia viruses (MLVs) able to replicate in murine cells. We demonstrate the spontaneous emergence of fully infectious ecotropic MLV in B6 mice with a range of distinct immune deficiencies affecting antibody production. These recombinant retroviruses establish infection of immunodeficient mouse colonies, and ultimately result in retrovirus-induced lymphomas. Notably, ERV activation in immunodeficient mice is prevented in husbandry conditions associated with reduced or absent intestinal microbiota. Our results shed light onto a previously unappreciated role for immunity in the control of ERVs and provide a potential mechanistic link between immune activation by microbial triggers and a range of pathologies associated with ERVs, including cancer.


Assuntos
Anticorpos Antivirais/biossíntese , Retrovirus Endógenos/fisiologia , Hospedeiro Imunocomprometido/imunologia , Ativação Viral , Criação de Animais Domésticos , Animais , Anticorpos Antivirais/imunologia , Transformação Celular Viral , Retrovirus Endógenos/genética , Retrovirus Endógenos/crescimento & desenvolvimento , Retrovirus Endógenos/imunologia , Feminino , Leucemia/virologia , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/crescimento & desenvolvimento , Vírus da Leucemia Murina/imunologia , Vírus da Leucemia Murina/fisiologia , Linfoma/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/genética , Recombinação Genética , Viremia/imunologia , Viremia/virologia
14.
PLoS One ; 7(9): e45171, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028824

RESUMO

Toll-like receptor (TLR)-dependent pathways control the activation of various immune cells and the production of cytokines and chemokines that are important in innate immune control of viruses, including mouse cytomegalovirus (MCMV). Here we report that upon MCMV infection wild-type and TLR7(-/-) male mice were more resistant than their female counterparts, while TLR9(-/-) male and female mice showed similar susceptibility. Interestingly, 36 h upon MCMV infection TLR9 mRNA expression was higher in male than in female mouse spleens. MCMV infection led to stronger reduction of marginal zone (MZ) B cells, and higher infiltration of plasmacytoid dendritic cells and neutrophils in wild-type male than female mice, while no such sex differences were observed in TLR9(-/-) mice. In accordance, the serum levels of KC and MIP-2, major neutrophil chemoattractants, were higher in wild-type, but not in TLR9(-/-), male versus female mice. Wild-type MCMV-infected female mice showed more severe liver inflammation, necrosis and steatosis compared to infected male mice. Our data demonstrate sex differences in susceptibility to MCMV infection, accompanied by a lower activation of the innate immune system in female mice, and can be attributed, at least in a certain degree, to the lower expression of TLR9 in female than male mice.


Assuntos
Infecções por Citomegalovirus/genética , Imunidade Inata , Glicoproteínas de Membrana/genética , Muromegalovirus/imunologia , Receptor 7 Toll-Like/genética , Receptor Toll-Like 9/genética , Animais , Linfócitos B/imunologia , Linfócitos B/virologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Células Dendríticas/imunologia , Células Dendríticas/virologia , Suscetibilidade a Doenças , Feminino , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Fígado/patologia , Fígado/virologia , Masculino , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/virologia , RNA Mensageiro/biossíntese , Fatores Sexuais , Baço/patologia , Baço/virologia , Receptor 7 Toll-Like/deficiência , Receptor 7 Toll-Like/imunologia , Receptor Toll-Like 9/deficiência , Receptor Toll-Like 9/imunologia
15.
PLoS Pathog ; 8(7): e1002812, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22829768

RESUMO

Group B Streptococcus (GBS) is a leading cause of invasive bacterial infections in human newborns and immune-compromised adults. The pore-forming toxin (PFT) ß hemolysin/cytolysin (ßh/c) is a major virulence factor for GBS, which is generally attributed to its cytolytic functions. Here we show ßh/c has immunomodulatory properties on macrophages at sub-lytic concentrations. ßh/c-mediated activation of p38 MAPK drives expression of the anti-inflammatory and immunosuppressive cytokine IL-10, and inhibits both IL-12 and NOS2 expression in GBS-infected macrophages, which are critical factors in host defense. Isogenic mutant bacteria lacking ßh/c fail to activate p38-mediated IL-10 production in macrophages and promote increased IL-12 and NOS2 expression. Furthermore, targeted deletion of p38 in macrophages increases resistance to invasive GBS infection in mice, associated with impaired IL-10 induction and increased IL-12 production in vivo. These data suggest p38 MAPK activation by ßh/c contributes to evasion of host defense through induction of IL-10 expression and inhibition of macrophage activation, a new mechanism of action for a PFT and a novel anti-inflammatory role for p38 in the pathogenesis of invasive bacterial infection. Our studies suggest p38 MAPK may represent a new therapeutic target to blunt virulence and improve clinical outcome of invasive GBS infection.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas Hemolisinas/metabolismo , Interleucina-10/biossíntese , Macrófagos/imunologia , Macrófagos/microbiologia , Infecções Estreptocócicas/imunologia , Streptococcus agalactiae/patogenicidade , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Imunidade Inata , Interleucina-12/biossíntese , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Camundongos , Óxido Nítrico Sintase/biossíntese , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae/imunologia , Streptococcus agalactiae/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética
16.
J Med Chem ; 55(11): 5642-6, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22578090
17.
Med Sci (Paris) ; 28(1): 96-102, 2012 Jan.
Artigo em Francês | MEDLINE | ID: mdl-22289837

RESUMO

Toll-like receptors (TLR) sense a variety of microbial products and play an important role in the mounting of innate and adaptive immune responses. TLR1 to TLR9 are common in mice and humans and recognize similar ligands in both species, with the exception of TLR8. Human TLR7 and TLR8 and mouse TLR7 detect viral single-stranded RNA and imidazoquinoline compounds, while mouse TLR8 not. Based on this discrepancy, for long time it was believed that mouse TLR8 is not functional and as a consequence the contribution of TLR8 to innate immunity remained poorly understood. Our recent studies revealed an important role for TLR8 in the regulation of TLR7-mediated autoimmunity in the mouse. This review illustrates our current understanding regarding the function of TLR8 and its potential for future clinical use for the treatment and/or prevention of various pathological conditions.


Assuntos
Glicoproteínas de Membrana/imunologia , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/fisiologia , Imunidade Adaptativa , Aminoquinolinas/farmacologia , Animais , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/imunologia , Ensaios Clínicos Fase I como Assunto , Citocinas/fisiologia , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica , Imiquimode , Imunidade Inata , Ligantes , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Modelos Imunológicos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , RNA Viral/imunologia , Transdução de Sinais/fisiologia , Especificidade da Espécie , Receptor 3 Toll-Like/fisiologia , Receptor 7 Toll-Like/agonistas , Receptor 7 Toll-Like/fisiologia , Receptor 8 Toll-Like/agonistas , Receptor 8 Toll-Like/genética , Viroses/imunologia
18.
Proc Natl Acad Sci U S A ; 108(6): 2372-7, 2011 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-21220319

RESUMO

The critical role of Toll-like receptors (TLRs) in mammalian host defense has been extensively explored in recent years. The capacity of about 10 TLRs to recognize conserved patterns on many bacterial and viral pathogens is remarkable. With so few receptors, cross-reactivity with self-tissue components often occurs. Previous studies have frequently assigned detrimental roles to TLRs, in particular to TLR2 and TLR4, in immune and cardiovascular disease. Using human and murine systems, we have investigated the consequence of TLR3 signaling in vascular disease. We compared the responses of human atheroma-derived smooth muscle cells (AthSMC) and control aortic smooth muscle cells (AoSMC) to various TLR ligands. AthSMC exhibited a specific increase in TLR3 expression and TLR3-dependent functional responses. Intriguingly, exposure to dsRNA in vitro and in vivo induced increased expression of both pro- and anti-inflammatory genes in vascular cells and tissues. Therefore, we sought to assess the contribution of TLR3 signaling in vivo in mechanical and hypercholesterolemia-induced arterial injury. Surprisingly, neointima formation in a perivascular collar-induced injury model was reduced by the systemic administration of the dsRNA analog Poly(I:C) in a TLR3-dependent manner. Furthermore, genetic deletion of TLR3 dramatically enhanced the development of elastic lamina damage after collar-induced injury. Accordingly, deficiency of TLR3 accelerated the onset of atherosclerosis in hypercholesterolemic ApoE(-/-) mice. Collectively, our data describe a protective role for TLR signaling in the vessel wall.


Assuntos
Artérias Carótidas/metabolismo , Doenças das Artérias Carótidas/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Transdução de Sinais , Receptor 3 Toll-Like/metabolismo , Animais , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/patologia , Feminino , Humanos , Hipercolesterolemia/metabolismo , Hipercolesterolemia/patologia , Indutores de Interferon/farmacologia , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Poli I-C/farmacologia , Receptor 3 Toll-Like/agonistas , Receptor 3 Toll-Like/genética
19.
Cell Immunol ; 267(1): 9-16, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21092943

RESUMO

Toll-like receptor 3 (TLR3) binds and signals in response to dsRNA and poly(I:C), a synthetic double stranded RNA analog. Activation of TLR3 triggers innate responses that may play a protective or detrimental role in viral infections or in immune-mediated inflammatory diseases through amplification of inflammation. Two monoclonal antibodies, CNTO4685 (rat anti-mouse TLR3) and CNTO5429 (CDRs from CNTO4685 grafted onto a mouse IgG1 scaffold) were generated and characterized. These mAbs bind the extracellular domain of mouse TLR3, inhibit poly(I:C)-induced activation of HEK293T cells transfected with mTLR3, and reduce poly(I:C)-induced production of CCL2 and CXCL10 by primary mouse embryonic fibroblasts. CNTO5429 decreased serum IL-6 and TNFα levels post-intraperitoneal poly(I:C) administration, demonstrating in vivo activity. In summary, specific anti-mTLR3 mAbs have been generated to assess TLR3 antagonism in mouse models of inflammation.


Assuntos
Anticorpos Monoclonais/imunologia , Poli I-C/imunologia , Receptor 3 Toll-Like/imunologia , Animais , Linhagem Celular , Células Cultivadas , Humanos , Inflamação/imunologia , Espaço Intracelular/imunologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptor 3 Toll-Like/genética
20.
J Clin Invest ; 120(10): 3651-62, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20811154

RESUMO

TLRs play an essential role in the induction of immune responses by detecting conserved molecular products of microorganisms. However, the function of TLR8 is largely unknown. In the current study, we investigated the role of TLR8 signaling in immunity in mice. We found that Tlr8(-/-) DCs overexpressed TLR7, were hyperresponsive to various TLR7 ligands, and showed stronger and faster NF-κB activation upon stimulation with the TLR7 ligand R848. Tlr8(-/-) mice showed splenomegaly, defective development of marginal zone (MZ) and B1 B cells, and increased serum levels of IgM and IgG2a. Furthermore, Tlr8(-/-) mice exhibited increased serum levels of autoantibodies against small nuclear ribonucleoproteins, ribonucleoprotein, and dsDNA and developed glomerulonephritis, whereas neither Tlr7(-/-) nor Tlr8(-/-)Tlr7(-/-) mice showed any of the phenotypes observed in Tlr8(-/-) mice. These data provide evidence for a pivotal role for mouse TLR8 in the regulation of mouse TLR7 expression and prevention of spontaneous autoimmunity.


Assuntos
Autoimunidade , Receptor 8 Toll-Like/fisiologia , Animais , Linfócitos B/fisiologia , Células Dendríticas/fisiologia , Macrófagos/fisiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/fisiologia , Receptor 8 Toll-Like/deficiência , Receptor 8 Toll-Like/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA