Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
PLoS Pathog ; 6(1): e1000722, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20090833

RESUMO

Macrophages are the first line of defense against pathogens. Upon infection macrophages usually produce high levels of proinflammatory mediators. However, macrophages can undergo an alternate polarization leading to a permissive state. In assessing global macrophage responses to the bacterial agent of Whipple's disease, Tropheryma whipplei, we found that T. whipplei induced M2 macrophage polarization which was compatible with bacterial replication. Surprisingly, this M2 polarization of infected macrophages was associated with apoptosis induction and a functional type I interferon (IFN) response, through IRF3 activation and STAT1 phosphorylation. Using macrophages from mice deficient for the type I IFN receptor, we found that this type I IFN response was required for T. whipplei-induced macrophage apoptosis in a JNK-dependent manner and was associated with the intracellular replication of T. whipplei independently of JNK. This study underscores the role of macrophage polarization in host responses and highlights the detrimental role of type I IFN during T. whipplei infection.


Assuntos
Apoptose/imunologia , Perfilação da Expressão Gênica , Interferon Tipo I/imunologia , Macrófagos/microbiologia , Transdução de Sinais/imunologia , Doença de Whipple/imunologia , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Expressão Gênica , Marcação In Situ das Extremidades Cortadas , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/metabolismo , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT1/metabolismo , Transfecção , Tropheryma/imunologia , Tropheryma/metabolismo , Doença de Whipple/genética , Doença de Whipple/metabolismo
10.
Blood ; 115(10): 1958-68, 2010 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-20068222

RESUMO

Small intestinal CD103(+) dendritic cells (DCs) have the selective ability to promote de novo generation of regulatory T cells via the production of retinoic acid (RA). Considering that aldehyde dehydrogenase (ALDH) activity controls the production of RA, we used a flow cytometry-based assay to measure ALDH activity at the single-cell level and to perform a comprehensive analysis of the RA-producing DC populations present in lymphoid and nonlymphoid mouse tissues. RA-producing DCs were primarily of the tissue-derived, migratory DC subtype and can be readily found in the skin and in the lungs as well as in their corresponding draining lymph nodes. The RA-producing skin-derived DCs were capable of triggering the generation of regulatory T cells, a finding demonstrating that the presence of RA-producing, tolerogenic DCs is not restricted to the intestinal tract as previously thought. Unexpectedly, the production of RA by skin DCs was restricted to CD103(-) DCs, indicating that CD103 expression does not constitute a "universal" marker for RA-producing mouse DCs. Finally, Toll-like receptor (TLR) triggering or the presence of a commensal microflora was not essential for the induction of ALDH activity in the discrete ALDH(+) DC subsets that characterize tissues constituting environmental interfaces.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Células de Langerhans/fisiologia , Linfonodos/fisiologia , Linfócitos T Reguladores/imunologia , Tretinoína/metabolismo , Aldeído Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Antígenos CD/metabolismo , Células Cultivadas , Cadeias alfa de Integrinas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Isoenzimas/metabolismo , Células de Langerhans/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Ativação Linfocitária/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3 , Retinal Desidrogenase , Pele , Linfócitos T Reguladores/metabolismo
11.
J Immunol ; 185(4): 2080-8, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20639488

RESUMO

Cross-talk between NK cells and dendritic cells (DCs) is critical for the potent therapeutic response to dsRNA, but the receptors involved remained controversial. We show in this paper that two dsRNAs, polyadenylic-polyuridylic acid and polyinosinic-polycytidylic acid [poly(I:C)], similarly engaged human TLR3, whereas only poly(I:C) triggered human RIG-I and MDA5. Both dsRNA enhanced NK cell activation within PBMCs but only poly(I:C) induced IFN-gamma. Although myeloid DCs (mDCs) were required for NK cell activation, induction of cytolytic potential and IFN-gamma production did not require contact with mDCs but was dependent on type I IFN and IL-12, respectively. Poly(I:C) but not polyadenylic-polyuridylic acid synergized with mDC-derived IL-12 for IFN-gamma production by acting directly on NK cells. Finally, the requirement of both TLR3 and Rig-like receptor (RLR) on mDCs and RLRs but not TLR3 on NK cells for IFN-gamma production was demonstrated using TLR3- and Cardif-deficient mice and human RIG-I-specific activator. Thus, we report the requirement of cotriggering TLR3 and RLR on mDCs and RLRs on NK cells for a pathogen product to induce potent innate cell activation.


Assuntos
RNA Helicases DEAD-box/metabolismo , Células Dendríticas/efeitos dos fármacos , Interferon gama/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Receptor 3 Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Relação Dose-Resposta a Droga , Humanos , Helicase IFIH1 Induzida por Interferon , Células Matadoras Naturais/citologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Células Mieloides/metabolismo , Poli A-U/farmacologia , Poli I-C/farmacologia , RNA de Cadeia Dupla/farmacologia , Receptores Imunológicos , Receptor 3 Toll-Like/genética , Transfecção
12.
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
13.
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
14.
J Exp Med ; 202(2): 321-31, 2005 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-16027240

RESUMO

Systemic autoimmune disease in humans and mice is characterized by loss of immunologic tolerance to a restricted set of self-nuclear antigens. Autoantigens, such as double-stranded (ds) DNA and the RNA-containing Smith antigen (Sm), may be selectively targeted in systemic lupus erythematosus because of their ability to activate a putative common receptor. Toll-like receptor 9 (TLR9), a receptor for CpG DNA, has been implicated in the activation of autoreactive B cells in vitro, but its role in promoting autoantibody production and disease in vivo has not been determined. We show that in TLR9-deficient lupus-prone mice, the generation of anti-dsDNA and antichromatin autoantibodies is specifically inhibited. Other autoantibodies, such as anti-Sm, are maintained and even increased in TLR9-deficient mice. In contrast, ablation of TLR3, a receptor for dsRNA, did not inhibit the formation of autoantibodies to either RNA- or DNA-containing antigens. Surprisingly, we found that despite the lack of anti-dsDNA autoantibodies in TLR9-deficient mice, there was no effect on the development of clinical autoimmune disease or nephritis. These results demonstrate a specific requirement for TLR9 in autoantibody formation in vivo and indicate a critical role for innate immune activation in autoimmunity.


Assuntos
Anticorpos Antinucleares/imunologia , Formação de Anticorpos/imunologia , Proteínas de Ligação a DNA/imunologia , Nefrite Lúpica/imunologia , Receptores de Superfície Celular/imunologia , Animais , Autoantígenos , Autoimunidade , Linfócitos B/imunologia , DNA/imunologia , Proteínas de Ligação a DNA/genética , Humanos , Imunidade Inata , Nefrite Lúpica/genética , Nefrite Lúpica/patologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Receptores de Superfície Celular/genética , Ribonucleoproteínas Nucleares Pequenas/imunologia , Receptor 3 Toll-Like , Receptor Toll-Like 9 , Receptores Toll-Like , Proteínas Centrais de snRNP
15.
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
16.
J Immunol ; 183(6): 3712-9, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19710451

RESUMO

TLR3 is known to respond to dsRNA from viruses, apoptotic cells, and/or necrotic cells. Dying cells are a rich source of ligands that can activate TLRs, such as TLR3. TLR3 expressed in the liver is likely to be a mediator of innate activation and inflammation in the liver. The importance of this function of TLR3 during acute hepatitis has not previously been fully explored. We used the mouse model of Con A-induced hepatitis and observed a novel role for TLR3 in hepatocyte damage in the absence of an exogenous viral stimulus. Interestingly, TLR3 expression in liver mononuclear cells and sinus endothelial cells was up-regulated after Con A injection and TLR3(-/-) mice were protected from Con A-induced hepatitis. Moreover, splenocytes from TLR3(-/-) mice proliferated less to Con A stimulation in the presence of RNA derived from damaged liver tissue compared with wild-type (WT) mice. To determine the relative contribution of TLR3 expression by hematopoietic cells or nonhematopoietic to liver damage during Con A-induced hepatitis, we generated bone marrow chimeric mice. TLR3(-/-) mice engrafted with WT hematopoietic cells were protected in a similar manner to WT mice reconstituted with TLR3(-/-) bone marrow, indicating that TLR3 signaling in both nonhematopoietic and hematopoietic cells plays an important role in mediating liver damage. In summary, our data suggest that TLR3 signaling is necessary for Con A-induced liver damage in vivo and that TLR3 regulates inflammation and the adaptive T cell immune response in the absence of viral infection.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/patologia , Inflamação/etiologia , Receptor 3 Toll-Like/fisiologia , Doença Aguda , Animais , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Concanavalina A , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Hepatócitos/patologia , Fígado/metabolismo , Fígado/patologia , Camundongos , Linfócitos T/imunologia , Receptor 3 Toll-Like/administração & dosagem , Receptor 3 Toll-Like/deficiência , Receptor 3 Toll-Like/genética
17.
Nat Med ; 10(12): 1366-73, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15558055

RESUMO

West Nile virus (WNV), a mosquito-borne single-stranded (ss)RNA flavivirus, causes human disease of variable severity. We investigated the involvement of Toll-like receptor (Tlr) 3, which recognizes viral double-stranded (ds)RNA, on WNV infection. Tlr3-deficient (Tlr3(-/-)) mice were more resistant to lethal WNV infection and had impaired cytokine production and enhanced viral load in the periphery, whereas in the brain, viral load, inflammatory responses and neuropathology were reduced compared to wild-type mice. Peripheral WNV infection led to a breakdown of the blood-brain barrier and enhanced brain infection in wild-type but not in Tlr3(-/-) mice, although both groups were equally susceptible upon intracerebroventricular administration of the virus. Tumor necrosis factor-alpha receptor 1 signaling is vital for blood-brain barrier compromise upon Tlr3 stimulation by dsRNA or WNV. Collectively, WNV infection leads to a Tlr3-dependent inflammatory response, which is involved in brain penetration of the virus and neuronal injury.


Assuntos
Barreira Hematoencefálica/virologia , Encéfalo/virologia , Encefalite/etiologia , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Vírus do Nilo Ocidental/metabolismo , Animais , Apoptose/fisiologia , Barreira Hematoencefálica/metabolismo , Encefalite/virologia , Imuno-Histoquímica , Inflamação/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Permeabilidade , Receptor 3 Toll-Like , Receptores Toll-Like , Fator de Necrose Tumoral alfa/metabolismo , Carga Viral
18.
Nat Med ; 8(8): 878-84, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12091878

RESUMO

The Lyme disease vaccine is based on the outer-surface lipoprotein (OspA) of the pathogen Borrelia burgdorferi, and 95% of vaccine recipients develop substantial titers of antibodies against OspA. Here, we identified seven individuals with very low antibody titers after vaccination (low responders). The macrophages of low responders produced less tumor necrosis factor-alpha and interleukin-6 after OspA stimulation and had lower cell-surface expression of Toll-like receptor (TLR) 1 as compared to normal cells, but normal expression of TLR2. TLRs activate innate responses to pathogens, and TLR2 recognizes lipoproteins and peptidoglycan (PGN). After OspA immunization, mice genetically deficient in either TLR2 (TLR2(-/-)) or TLR1 (TLR1(-/-)) produced low titers of antibodies against OspA. Notably, macrophages from TLR2(-/-) mice were unresponsive to OspA and PGN, whereas those from TLR1(-/-) mice responded normally to PGN but not to OspA. These data indicate that TLR1 and TLR2 are required for lipoprotein recognition and that defects in the TLR1/2 signaling pathway may account for human hyporesponsiveness to OspA vaccination.


Assuntos
Anticorpos Antibacterianos/sangue , Antígenos de Superfície/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Borrelia burgdorferi/imunologia , Proteínas de Drosophila , Vacinas contra Doença de Lyme/imunologia , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Vacinas Bacterianas , Borrelia burgdorferi/química , Separação Celular , Células Cultivadas , Humanos , Interleucinas/metabolismo , Lipoproteínas/imunologia , Lipoproteínas/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Receptores de Superfície Celular/genética , Transdução de Sinais , Receptor 1 Toll-Like , Receptor 2 Toll-Like , Receptores Toll-Like
19.
Nature ; 433(7028): 887-92, 2005 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-15711573

RESUMO

Cross-presentation of cell-associated antigens plays an important role in regulating CD8+ T cell responses to proteins that are not expressed by antigen-presenting cells (APCs). Dendritic cells are the principal cross-presenting APCs in vivo and much progress has been made in elucidating the pathways that allow dendritic cells to capture and process cellular material. However, little is known about the signals that determine whether such presentation ultimately results in a cytotoxic T cell (CTL) response (cross-priming) or in CD8+ T cell inactivation (cross-tolerance). Here we describe a mechanism that promotes cross-priming during viral infections. We show that murine CD8alpha+ dendritic cells are activated by double-stranded (ds)RNA present in virally infected cells but absent from uninfected cells. Dendritic cell activation requires phagocytosis of infected material, followed by signalling through the dsRNA receptor, toll-like receptor 3 (TLR3). Immunization with virus-infected cells or cells containing synthetic dsRNA leads to a striking increase in CTL cross-priming against cell-associated antigens, which is largely dependent on TLR3 expression by antigen-presenting cells. Thus, TLR3 may have evolved to permit cross-priming of CTLs against viruses that do not directly infect dendritic cells.


Assuntos
Apresentação de Antígeno/imunologia , Infecções por Cardiovirus/imunologia , Apresentação Cruzada/imunologia , Glicoproteínas de Membrana/metabolismo , Receptores de Superfície Celular/metabolismo , Linfócitos T Citotóxicos/imunologia , Animais , Chlorocebus aethiops , Células Dendríticas/imunologia , Vírus da Encefalomiocardite/imunologia , Vírus da Encefalomiocardite/fisiologia , Endossomos/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Poli I-C/imunologia , Poli I-C/metabolismo , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/imunologia , RNA de Cadeia Dupla/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Superfície Celular/genética , Receptor 3 Toll-Like , Receptores Toll-Like , Células Vero
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA