Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 22(8): 947-957, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34239121

RESUMO

One of most challenging issues in tumor immunology is a better understanding of the dynamics in the accumulation of myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment (TIME), as this would lead to the development of new cancer therapeutics. Here, we show that translationally controlled tumor protein (TCTP) released by dying tumor cells is an immunomodulator crucial to full-blown MDSC accumulation in the TIME. We provide evidence that extracellular TCTP mediates recruitment of the polymorphonuclear MDSC (PMN-MDSC) population in the TIME via activation of Toll-like receptor-2. As further proof of principle, we show that inhibition of TCTP suppresses PMN-MDSC accumulation and tumor growth. In human cancers, we find an elevation of TCTP and an inverse correlation of TCTP gene dosage with antitumor immune signatures and clinical prognosis. This study reveals the hitherto poorly understood mechanism of the MDSC dynamics in the TIME, offering a new rationale for cancer immunotherapy.


Assuntos
Biomarcadores Tumorais/metabolismo , Quimiocina CXCL1/metabolismo , Neoplasias Colorretais/imunologia , Células Supressoras Mieloides/imunologia , Receptor 2 Toll-Like/imunologia , Microambiente Tumoral/imunologia , Alarminas/genética , Alarminas/metabolismo , Animais , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Imunoterapia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células RAW 264.7 , Proteína Tumoral 1 Controlada por Tradução
2.
Cell ; 159(7): 1563-77, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25525875

RESUMO

The mechanism by which cells undergo death determines whether dying cells trigger inflammatory responses or remain immunologically silent. Mitochondria play a central role in the induction of cell death, as well as in immune signaling pathways. Here, we identify a mechanism by which mitochondria and downstream proapoptotic caspases regulate the activation of antiviral immunity. In the absence of active caspases, mitochondrial outer membrane permeabilization by Bax and Bak results in the expression of type I interferons (IFNs). This induction is mediated by mitochondrial DNA-dependent activation of the cGAS/STING pathway and results in the establishment of a potent state of viral resistance. Our results show that mitochondria have the capacity to simultaneously expose a cell-intrinsic inducer of the IFN response and to inactivate this response in a caspase-dependent manner. This mechanism provides a dual control, which determines whether mitochondria initiate an immunologically silent or a proinflammatory type of cell death.


Assuntos
Apoptose , Caspases/metabolismo , Interferon Tipo I/metabolismo , Transdução de Sinais , Animais , DNA Mitocondrial/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Interferon Tipo I/imunologia , Camundongos , Camundongos Knockout , Viroses/imunologia
3.
Proc Natl Acad Sci U S A ; 120(24): e2305245120, 2023 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-37276392

RESUMO

The activation and expansion of T cells that recognize cancer cells is an essential aspect to antitumor immunity. Tumors may escape destruction by the immune system through ectopic expression of inhibitory immune ligands typically exemplified by the PD-L1/PD-1 pathway. Here, we reveal another facet of tumor evasion from T cell surveillance. By secretome profiling of necrotic tumor cells, we identified an oncometabolite spermidine as a unique inhibitor of T cell receptor (TCR) signaling. Mechanistically, spermidine causes the downregulation of the plasma membrane cholesterol levels, resulting in the suppression of TCR clustering. Using syngeneic mouse models, we show that spermidine is abundantly detected in the tumor immune microenvironment (TIME) and that administration of the polyamine synthesis inhibitor effectively enhanced CD8+ T cell-dependent antitumor responses. Further, the combination of the polyamine synthesis inhibitor with anti-PD-1 immune checkpoint antibody resulted in a much stronger antitumor immune response. This study reveals an aspect of immunosuppressive TIME, wherein spermidine functions as a metabolic T cell checkpoint that may offer a unique approach for promoting tumor immunotherapy.


Assuntos
Antineoplásicos , Neoplasias , Animais , Camundongos , Espermidina/farmacologia , Espermidina/metabolismo , Linfócitos T CD8-Positivos , Neoplasias/metabolismo , Antineoplásicos/farmacologia , Imunoterapia/métodos , Receptores de Antígenos de Linfócitos T/metabolismo , Microambiente Tumoral , Linhagem Celular Tumoral , Antígeno B7-H1/metabolismo
4.
Immunity ; 45(2): 319-32, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27521268

RESUMO

Interferon regulatory factor-5 (IRF5), a transcription factor critical for the induction of innate immune responses, contributes to the pathogenesis of the autoimmune disease systemic lupus erythematosus (SLE) in humans and mice. Lyn, a Src family kinase, is also implicated in human SLE, and Lyn-deficient mice develop an SLE-like disease. Here, we found that Lyn physically interacted with IRF5 to inhibit ubiquitination and phosphorylation of IRF5 in the TLR-MyD88 pathway, thereby suppressing the transcriptional activity of IRF5 in a manner independent of Lyn's kinase activity. Conversely, Lyn did not inhibit NF-κB signaling, another major branch downstream of MyD88. Monoallelic deletion of Irf5 alleviated the hyperproduction of cytokines in TLR-stimulated Lyn(-/-) dendritic cells and the development of SLE-like symptoms in Lyn(-/-) mice. Our results reveal a role for Lyn as a specific suppressor of the TLR-MyD88-IRF5 pathway and illustrate the importance of fine-tuning IRF5 activity for the maintenance of immune homeostasis.


Assuntos
Autoimunidade , Células Dendríticas/imunologia , Fatores Reguladores de Interferon/metabolismo , Lúpus Eritematoso Sistêmico/imunologia , Quinases da Família src/metabolismo , Animais , Células Cultivadas , Citocinas/metabolismo , Humanos , Tolerância Imunológica , Imunidade Inata , Fatores Reguladores de Interferon/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Fosforilação , Ligação Proteica , Transdução de Sinais , Receptores Toll-Like/metabolismo , Ativação Transcricional , Ubiquitinação , Quinases da Família src/genética
5.
Genes Cells ; 28(3): 202-210, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36550748

RESUMO

High-mobility group box 1 (HMGB1) is a nucleotide-binding chromatin protein that has also been characterized as a prototypical damage-associate molecular pattern. It triggers inflammatory responses upon release from damaged or dying cells. In fact, HMGB1 has been linked to the induction of many inflammatory diseases through immune cell activation including neutrophil recruitment. In this study, we examined the impact of HMGB1-binding inhibitory oligodeoxynucleotide (ISM ODN) on the development of hepatitis using a murine model of the disease. Our results indicate that ISM ODN effectively suppresses pathological features of hepatitis, including neutrophil accumulation. This study therefore may offer clinical insight into the treatment of hepatitis and possibly other inflammatory diseases.


Assuntos
Proteína HMGB1 , Hepatite , Camundongos , Animais , Proteína HMGB1/metabolismo , Oligodesoxirribonucleotídeos/farmacologia , Modelos Animais de Doenças , Infiltração de Neutrófilos
6.
Nat Immunol ; 13(7): 659-66, 2012 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-22610141

RESUMO

Although the mechanisms by which innate pathogen-recognition receptors enhance adaptive immune responses are increasingly well understood, whether signaling events from distinct classes of receptors affect each other in modulating adaptive immunity remains unclear. We found here that the activation of cytosolic RIG-I-like receptors (RLRs) resulted in the selective suppression of transcription of the gene encoding the p40 subunit of interleukin 12 (Il12b) that was effectively induced by the activation of Toll-like receptors (TLRs). The RLR-activated transcription factor IRF3 bound dominantly, relative to IRF5, to the Il12b promoter, where it interfered with the TLR-induced assembly of a productive transcription-factor complex. The activation of RLRs in mice attenuated TLR-induced responses of the T helper type 1 cell (T(H)1 cell) and interleukin 17-producing helper T cell (T(H)17 cell) subset types and, consequently, viral infection of mice caused death at sublethal doses of bacterial infection. The innate immune receptor cross-interference we describe may have implications for infection-associated clinical episodes.


Assuntos
Transdução de Sinais/imunologia , Linfócitos T/imunologia , Receptores Toll-Like/imunologia , Sequência de Aminoácidos , Animais , Infecções Bacterianas/imunologia , Células Cultivadas , Regulação da Expressão Gênica/imunologia , Fator Regulador 3 de Interferon/metabolismo , Fatores Reguladores de Interferon/metabolismo , Subunidade p40 da Interleucina-12/metabolismo , Macrófagos Peritoneais/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Células Th1/imunologia , Células Th17/imunologia , Fatores de Transcrição/metabolismo , Viroses/imunologia
7.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33443188

RESUMO

Dysregulation of inflammatory cytokines in keratinocytes promote the pathogenesis of the skin inflammation, such as allergic contact dermatitis (ACD). High-mobility group box 1 protein (HMGB1) has been implicated in the promotion of skin inflammation upon its extracellular release as a damage-associated molecular pattern molecule. However, whether and how HMGB1 in keratinocytes contributes to ACD and other skin disorders remain elusive. In this study, we generated conditional knockout mice in which the Hmgb1 gene is specifically deleted in keratinocytes, and examined its role in ACD models. Interestingly, the mutant mice showed exacerbated skin inflammation, accompanied by increased ear thickening in 2,4-dinitrofluorobenezene-induced ACDs. The mRNA expression of interleukin-24 (IL-24), a cytokine known to critically contribute to ACD pathogenesis, was elevated in skin lesions of the mutant mice. As with constitutively expressed, IL-4-induced Il24 mRNA, expression was also augmented in the Hmgb1-deficient keratinocytes, which would account for the exacerbation of ACD in the mutant mice. Mechanistically, we observed an increased binding of trimethyl histone H3 (lys4) (H3K4me3), a hallmark of transcriptionally active genes, to the promoter region of the Il24 gene in the hmgb1-deficient cells. Thus, the nuclear HMGB1 is a critical "gate keeper" in that the dermal homeostasis is contingent to its function in chromatin remodeling. Our study revealed a facet of nuclear HMGB1, namely its antiinflammatory function in keratinocytes for the skin homeostasis.


Assuntos
Montagem e Desmontagem da Cromatina , Dermatite Alérgica de Contato/metabolismo , Proteína HMGB1/metabolismo , Histonas/metabolismo , Interleucinas/metabolismo , Queratinócitos/metabolismo , Animais , Dermatite Alérgica de Contato/genética , Dermatite Alérgica de Contato/prevenção & controle , Dinitrofluorbenzeno/toxicidade , Modelos Animais de Doenças , Orelha/patologia , Deleção de Genes , Regulação da Expressão Gênica/genética , Proteína HMGB1/deficiência , Proteína HMGB1/genética , Inflamação/genética , Inflamação/metabolismo , Interleucina-4/farmacologia , Interleucinas/genética , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas , Pele/imunologia , Pele/metabolismo , Pele/patologia , Quimeras de Transplante
8.
Int Immunol ; 33(12): 841-846, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34357403

RESUMO

As clinically demonstrated by the success of immunotherapies to improve survival outcomes, tumors are known to gain a survival advantage by circumventing immune surveillance. A defining feature of this is the creation and maintenance of a tumor immune microenvironment (TIME) that directly and indirectly alters the host's immunologic signaling pathways through a variety of mechanisms. Tumor-intrinsic mechanisms that instruct the formation and maintenance of the TIME have been an area of intensive study, such as the identification and characterization of soluble factors actively and passively released by tumor cells that modulate immune cell function. In particular, damage-associated molecular pattern (DAMP) molecules typically released by necrotic tumor cells are recognized by innate immune receptors such as Toll-like receptors (TLRs) and stimulate immune cells within TIME. Given their broad and potent effects on the immune system, a better understanding for how DAMP and TLR interactions sculpt the TIME to favor tumor growth would identify new strategies and approaches for cancer immunotherapy.


Assuntos
Neoplasias/imunologia , Receptores Toll-Like/imunologia , Animais , Humanos , Microambiente Tumoral/imunologia
9.
Proc Natl Acad Sci U S A ; 116(47): 23653-23661, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31694883

RESUMO

The activation of innate immune receptors by pathogen-associated molecular patterns (PAMPs) is central to host defense against infections. On the other hand, these receptors are also activated by immunogenic damage-associated molecular patterns (DAMPs), typically released from dying cells, and the activation can evoke chronic inflammatory or autoimmune disorders. One of the best known receptors involved in the immune pathogenesis is Toll-like receptor 7 (TLR7), which recognizes RNA with single-stranded structure. However, the causative DAMP RNA(s) in the pathogenesis has yet to be identified. Here, we first developed a chemical compound, termed KN69, that suppresses autoimmunity in several established mouse models. A subsequent search for KN69-binding partners led to the identification of U11 small nuclear RNA (U11snRNA) as a candidate DAMP RNA involved in TLR7-induced autoimmunity. We then showed that U11snRNA robustly activated the TLR7 pathway in vitro and induced arthritis disease in vivo. We also found a correlation between high serum level of U11snRNA and autoimmune diseases in human subjects and established mouse models. Finally, by revealing the structural basis for U11snRNA's ability to activate TLR7, we developed more potent TLR7 agonists and TLR7 antagonists, which may offer new therapeutic approaches for autoimmunity or other immune-driven diseases. Thus, our study has revealed a hitherto unknown immune function of U11snRNA, providing insight into TLR7-mediated autoimmunity and its potential for further therapeutic applications.


Assuntos
Glicoproteínas de Membrana/agonistas , RNA Nuclear Pequeno/imunologia , Receptor 7 Toll-Like/agonistas , Adulto , Alarminas/química , Animais , Artrite Reumatoide/sangue , Artrite Reumatoide/imunologia , Doenças Autoimunes/sangue , Doenças Autoimunes/imunologia , Sequência de Bases , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Imunossupressores/síntese química , Imunossupressores/farmacologia , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/imunologia , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Pessoa de Meia-Idade , RNA/imunologia , RNA/metabolismo , Ribonucleoproteínas Nucleares Pequenas/química , Ribonucleoproteínas Nucleares Pequenas/imunologia , Análise de Sequência de RNA , Receptor 7 Toll-Like/deficiência , Adulto Jovem
10.
Cancer Sci ; 112(7): 2578-2591, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33570784

RESUMO

The signal-transducing innate receptors represent classes of pattern recognition receptors (PRRs) that play crucial roles in the first line of the host defense against infections by the recognition of pathogen-derived molecules. Because of their poorly discriminative nature compared with antigen receptors of the adaptive immune system, they also recognize endogenous molecules and evoke immune responses without infection, resulting in the regulation of tumor immunity. Therefore, PRRs may be promising targets for effective cancer immunotherapy, either by activating or inhibiting them. Here, we summarize our current knowledge of signal-transducing PRRs in the regulation of tumor immunity.


Assuntos
Neoplasias/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Transdução de Sinais/imunologia , Animais , Membrana Celular/metabolismo , Citoplasma/metabolismo , Humanos , Imunidade Inata , Imunoterapia/tendências , Neoplasias/terapia , Receptores de Reconhecimento de Padrão/metabolismo , Microambiente Tumoral/imunologia
11.
Proc Natl Acad Sci U S A ; 115(20): 5253-5258, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29712834

RESUMO

IFN regulatory factor 3 (IRF3) is a transcription regulator of cellular responses in many cell types that is known to be essential for innate immunity. To confirm IRF3's broad role in immunity and to more fully discern its role in various cellular subsets, we engineered Irf3-floxed mice to allow for the cell type-specific ablation of Irf3 Analysis of these mice confirmed the general requirement of IRF3 for the evocation of type I IFN responses in vitro and in vivo. Furthermore, immune cell ontogeny and frequencies of immune cell types were unaffected when Irf3 was selectively inactivated in either T cells or B cells in the mice. Interestingly, in a model of lipopolysaccharide-induced septic shock, selective Irf3 deficiency in myeloid cells led to reduced levels of type I IFN in the sera and increased survival of these mice, indicating the myeloid-specific, pathogenic role of the Toll-like receptor 4-IRF3 type I IFN axis in this model of sepsis. Thus, Irf3-floxed mice can serve as useful tool for further exploring the cell type-specific functions of this transcription factor.


Assuntos
Imunidade Inata/imunologia , Inflamação/imunologia , Fator Regulador 3 de Interferon/metabolismo , Células Mieloides/imunologia , Linfócitos T/imunologia , Animais , Regulação da Expressão Gênica , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/patologia , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Knockout , Células Mieloides/metabolismo , Células Mieloides/patologia , Transdução de Sinais , Linfócitos T/metabolismo , Linfócitos T/patologia
12.
Nat Immunol ; 9(1): 34-41, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18059273

RESUMO

Interleukin-12 (IL-12) and interferon-gamma (IFN-gamma) drive T helper type 1 (T(H)1) differentiation, but the mechanisms underlying the regulation of the complicated gene networks involved in this differentiation are not fully understood. Here we show that the IFN-gamma-induced transcription factor IRF1 was essential in T(H)1 differentiation by acting on Il12rb1, the gene encoding the IL-12 receptor beta1 subunit (IL-12Rbeta1). IRF1 directly interacted with and activated the Il12rb1 promoter in CD4+ T cells. Notably, the IRF1-dependent induction of IL-12Rbeta1 was essential for IFN-gamma-IL-12 signaling but was dispensable for IL-23-IL-17 signaling. Because both IL-12 and IL-23 bind to and transmit signals through IL-12Rbeta1, our data suggest that distinct thresholds of IL-12Rbeta1 expression are required for T(H)1 versus T(H)-17 differentiation.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fator Regulador 1 de Interferon/fisiologia , Interferon gama/imunologia , Interleucina-12/fisiologia , Células Th1/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Diferenciação Celular , Células Cultivadas , Fator Regulador 1 de Interferon/genética , Interleucina-17/fisiologia , Interleucina-23/fisiologia , Ativação Linfocitária , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas , Receptores de Interleucina-12/fisiologia , Transdução de Sinais , Células Th1/citologia
13.
Proc Natl Acad Sci U S A ; 114(38): 10178-10183, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28878025

RESUMO

The commensal microbiota within the gastrointestinal tract is essential in maintaining homeostasis. Indeed, dysregulation in the repertoire of microbiota can result in the development of intestinal immune-inflammatory diseases. Further, this immune regulation by gut microbiota is important systemically, impacting health and disease of organ systems beyond the local environment of the gut. What has not been explored is how distant organs might in turn shape the microbiota via microbe-targeted molecules. Here, we provide evidence that surfactant protein D (SP-D) synthesized in the gallbladder and delivered into intestinal lumen binds selectively to species of gut commensal bacteria. SP-D-deficient mice manifest intestinal dysbiosis and show a susceptibility to dextran sulfate sodium-induced colitis. Further, fecal transfer from SP-D-deficient mice to wild-type, germ-free mice conveyed colitis susceptibility. Interestingly, colitis caused a notable increase in Sftpd gene expression in the gallbladder, but not in the lung, via the activity of glucocorticoids produced in the liver. These findings describe a unique mechanism of interorgan regulation of intestinal immune homeostasis by SP-D with potential clinical implications such as cholecystectomy.


Assuntos
Colite/metabolismo , Vesícula Biliar/metabolismo , Microbioma Gastrointestinal , Proteína D Associada a Surfactante Pulmonar/metabolismo , Animais , Colite/microbiologia , Fatores de Transcrição Forkhead/metabolismo , Glucocorticoides/biossíntese , Homeostase , Mucosa Intestinal/imunologia , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Simbiose , Linfócitos T Reguladores/metabolismo
14.
Nature ; 500(7461): 232-6, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-23842501

RESUMO

Manipulation of the gut microbiota holds great promise for the treatment of inflammatory and allergic diseases. Although numerous probiotic microorganisms have been identified, there remains a compelling need to discover organisms that elicit more robust therapeutic responses, are compatible with the host, and can affect a specific arm of the host immune system in a well-controlled, physiological manner. Here we use a rational approach to isolate CD4(+)FOXP3(+) regulatory T (Treg)-cell-inducing bacterial strains from the human indigenous microbiota. Starting with a healthy human faecal sample, a sequence of selection steps was applied to obtain mice colonized with human microbiota enriched in Treg-cell-inducing species. From these mice, we isolated and selected 17 strains of bacteria on the basis of their high potency in enhancing Treg cell abundance and inducing important anti-inflammatory molecules--including interleukin-10 (IL-) and inducible T-cell co-stimulator (ICOS)--in Treg cells upon inoculation into germ-free mice. Genome sequencing revealed that the 17 strains fall within clusters IV, XIVa and XVIII of Clostridia, which lack prominent toxins and virulence factors. The 17 strains act as a community to provide bacterial antigens and a TGF-ß-rich environment to help expansion and differentiation of Treg cells. Oral administration of the combination of 17 strains to adult mice attenuated disease in models of colitis and allergic diarrhoea. Use of the isolated strains may allow for tailored therapeutic manipulation of human immune disorders.


Assuntos
Clostridium/imunologia , Metagenoma/imunologia , Linfócitos T Reguladores/fisiologia , Adulto , Animais , Proliferação de Células , Clostridium/classificação , Clostridium/genética , Colite/microbiologia , Colite/patologia , Colo/imunologia , Colo/microbiologia , Modelos Animais de Doenças , Fezes/microbiologia , Vida Livre de Germes , Humanos , Proteína Coestimuladora de Linfócitos T Induzíveis/metabolismo , Interleucina-10/metabolismo , Masculino , Metagenoma/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos SCID , RNA Ribossômico 16S/genética , Ratos , Ratos Endogâmicos F344 , Linfócitos T Reguladores/citologia
15.
Proc Natl Acad Sci U S A ; 113(14): 3844-9, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-27001836

RESUMO

Cellular components released into the external milieu as a result of cell death and sensed by the body are generally termed damage-associated molecular patterns (DAMPs). Although DAMPs are conventionally thought to be protective to the host by evoking inflammatory responses important for immunity and wound repair, there is the prevailing notion that dysregulated release of DAMPs can also underlie or exacerbate disease development. However, the critical issue for how resultant DAMP-mediated responses are regulated has heretofore not been fully addressed. In the present study, we identify prostaglandin E2 (PGE2) as a DAMP that negatively regulates immune responses. We show that the production of PGE2 is augmented under cell death-inducing conditions via the transcriptional induction of the cyclooxygenase 2 (COX2) gene and that cell-released PGE2 suppresses the expression of genes associated with inflammation, thereby limiting the cell's immunostimulatory activities. Consistent with this, inhibition of the PGE2 synthesis pathway potentiates the inflammation induced by dying cells. We also provide in vivo evidence for a protective role of PGE2 released upon acetaminophen-induced liver injury as well as a pathogenic role for PGE2 during tumor cell growth. Our study places this classically known lipid mediator in an unprecedented context-that is, an inhibitory DAMP vis-à-vis activating DAMPs, which may have translational implications for designing more effective therapeutic regimens for inflammation-associated diseases.


Assuntos
Alarminas/metabolismo , Morte Celular/imunologia , Ciclo-Oxigenase 2/biossíntese , Dinoprostona/metabolismo , Inflamação/patologia , Acetaminofen/efeitos adversos , Animais , Morte Celular/fisiologia , Linhagem Celular Tumoral , Doença Hepática Induzida por Substâncias e Drogas/imunologia , Células HeLa , Humanos , Inflamação/imunologia , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos C57BL
16.
Proc Natl Acad Sci U S A ; 113(49): 14097-14102, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27872290

RESUMO

Tumor metastasis is the cause of most cancer deaths. Although metastases can form in multiple end organs, the liver is recognized as a highly permissive organ. Nevertheless, there is evidence for immune cell-mediated mechanisms that function to suppress liver metastasis by certain tumors, although the underlying mechanisms for the suppression of metastasis remain elusive. Here, we show that Dectin-2, a C-type lectin receptor (CLR) family of innate receptors, is critical for the suppression of liver metastasis of cancer cells. We provide evidence that Dectin-2 functions in resident macrophages in the liver, known as Kupffer cells, to mediate the uptake and clearance of cancer cells. Interestingly, Kupffer cells are selectively endowed with Dectin-2-dependent phagocytotic activity, with neither bone marrow-derived macrophages nor alveolar macrophages showing this potential. Concordantly, subcutaneous primary tumor growth and lung metastasis are not affected by the absence of Dectin-2. In addition, macrophage C-type lectin, a CLR known to be complex with Dectin-2, also contributes to the suppression of liver metastasis. Collectively, these results highlight the hitherto poorly understood mechanism of Kupffer cell-mediated control of metastasis that is mediated by the CLR innate receptor family, with implications for the development of anticancer therapy targeting CLRs.


Assuntos
Células de Kupffer/fisiologia , Lectinas Tipo C/metabolismo , Neoplasias Hepáticas Experimentais/secundário , Metástase Neoplásica/imunologia , Fagocitose , Animais , Linhagem Celular Tumoral , Humanos , Lectinas Tipo C/genética , Camundongos Endogâmicos C57BL , Receptores Imunológicos/metabolismo
17.
Proc Natl Acad Sci U S A ; 113(5): 1351-6, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26787880

RESUMO

Blunting immunopathology without abolishing host defense is the foundation for safe and effective modulation of infectious and autoimmune diseases. Sphingosine 1-phosphate receptor 1 (S1PR1) agonists are effective in treating infectious and multiple autoimmune pathologies; however, mechanisms underlying their clinical efficacy are yet to be fully elucidated. Here, we uncover an unexpected mechanism of convergence between S1PR1 and interferon alpha receptor 1 (IFNAR1) signaling pathways. Activation of S1PR1 signaling by pharmacological tools or endogenous ligand sphingosine-1 phosphate (S1P) inhibits type 1 IFN responses that exacerbate numerous pathogenic conditions. Mechanistically, S1PR1 selectively suppresses the type I IFN autoamplification loop in plasmacytoid dendritic cells (pDCs), a specialized DC subset, for robust type I IFN release. S1PR1 agonist suppression is pertussis toxin-resistant, but inhibited by an S1PR1 C-terminal-derived transactivating transcriptional activator (Tat)-fusion peptide that blocks receptor internalization. S1PR1 agonist treatment accelerates turnover of IFNAR1, suppresses signal transducer and activator of transcription 1 (STAT1) phosphorylation, and down-modulates total STAT1 levels, thereby inactivating the autoamplification loop. Inhibition of S1P-S1PR1 signaling in vivo using the selective antagonist Ex26 significantly elevates IFN-α production in response to CpG-A. Thus, multiple lines of evidence demonstrate that S1PR1 signaling sets the sensitivity of pDC amplification of IFN responses, thereby blunting pathogenic immune responses. These data illustrate a lipid G-protein coupled receptor (GPCR)-IFNAR1 regulatory loop that balances effective and detrimental immune responses and elevated endogenous S1PR1 signaling. This mechanism will likely be advantageous in individuals subject to a range of inflammatory conditions.


Assuntos
Células Dendríticas/metabolismo , Interferon-alfa/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Receptores de Lisoesfingolipídeo/fisiologia , Animais , Camundongos , Camundongos Knockout , Proteólise , Receptor de Interferon alfa e beta/genética
18.
Cancer Sci ; 109(9): 2687-2696, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29981256

RESUMO

Recent years have seen a number of regulatory approvals for immune oncology or immunotherapies based on their ability to enhance antitumor immune responses. Nevertheless, the majority of patients remain refractory to these treatments; hence, new therapies that augment current immunotherapies are required. Innate immune receptors that recognize nucleic acids are potent activators of subsequent T-cell responses and, as a result, can evoke potent antitumor immune responses. Herein, we present a novel compound N-{3-[(1,4'-bipiperidin)-1'-yl]propyl}-6-[4-(4-methylpiperazin-1-yl)phenyl]picolinamide (SINCRO; STING-mediated interferon-inducing and cytotoxic reagent, original) as an anticancer drug that activates the cytosolic DNA-sensing STING (stimulator of interferon genes) signaling pathway leading to the induction of type I interferon (IFN) genes. Indeed, IFN-ß gene induction by SINCRO is abolished in STING-deficient cells. In addition to its IFN-inducing activity, SINCRO shows STING-independent cytotoxic activity against cancer cells. SINCRO does not evoke DNA double-strand break or caspase-3 cleavage. Thus, SINCRO induces cell death in a method different from conventional apoptosis-inducing pathways. Finally, we provide evidence that giving SINCRO significantly attenuates in vivo tumor growth by both type I IFN-dependent and independent mechanisms. Thus, SINCRO is an attractive anticancer compound with dual function in that it evokes type I IFN response to promote antitumor immunity as well as inducing tumor cell death. SINCRO may provide a new platform for the development of drugs for effective cancer therapy.


Assuntos
Amidas/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Interferon beta/biossíntese , Proteínas de Membrana/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Ácidos Picolínicos/farmacologia , Piperidinas/farmacologia , Células 3T3 , Amidas/química , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Células HEK293 , Células HeLa , Humanos , Interferon beta/genética , Interferon beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/patologia , Ácidos Picolínicos/química , Piperidinas/química , Transdução de Sinais/efeitos dos fármacos
19.
Proc Natl Acad Sci U S A ; 112(41): 12770-5, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26420876

RESUMO

The regulation of intestinal homeostasis by the immune system involves the dynamic interplay between gut commensal microbiota and resident immune cells. It is well known that a large and diverse lymphocyte antigen receptor repertoire enables the immune system to recognize and respond to a wide range of invading pathogens. There is also an emerging appreciation for a critical role the T-cell receptor (TCR) repertoire serves in the maintenance of peripheral tolerance by regulatory T cells (Tregs). Nevertheless, how the diversity of the TCR repertoire in Tregs affects intestinal homeostasis remains unknown. To address this question, we studied mice whose T cells express a restricted TCR repertoire. We observed the development of spontaneous colitis, accompanied by the induction of T-helper type 17 cells in the colon that is driven by gut commensal microbiota. We provide further evidence that a restricted TCR repertoire causes a loss of tolerogenicity to microbiota, accompanied by a paucity of peripherally derived, Helios(-) Tregs and hyperactivation of migratory dendritic cells. These results thus reveal a new facet of the TCR repertoire in which Tregs require a diverse TCR repitoire for intestinal homeostasis, suggesting an additional driving force in the evolutional significance of the TCR repertoire.


Assuntos
Movimento Celular/imunologia , Colo/imunologia , Microbiota/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Movimento Celular/genética , Colo/microbiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Células Dendríticas/imunologia , Camundongos , Camundongos Mutantes , Receptores de Antígenos de Linfócitos T/genética , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia
20.
Proc Natl Acad Sci U S A ; 112(49): 15136-41, 2015 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-26598674

RESUMO

Systemic sclerosis (SSc) is a multisystem autoimmune disorder with clinical manifestations resulting from tissue fibrosis and extensive vasculopathy. A potential disease susceptibility gene for SSc is IFN regulatory factor 5 (IRF5), whose SNP is associated with milder clinical manifestations; however, the underlying mechanisms of this association remain elusive. In this study we examined IRF5-deficient (Irf5(-/-)) mice in the bleomycin-treated SSc murine model. We show that dermal and pulmonary fibrosis induced by bleomycin is attenuated in Irf5(-/-) mice. Interestingly, we find that multiple SSc-associated events, such as fibroblast activation, inflammatory cell infiltration, endothelial-to-mesenchymal transition, vascular destabilization, Th2/Th17 skewed immune polarization, and B-cell activation, are suppressed in these mice. We further provide evidence that IRF5, activated by Toll-like receptor 4 (TLR4), binds to the promoters of various key genes involved in SSc disease pathology. These observations are congruent with the high level of expression of IRF5, TLR4, and potential endogenous TLR4 ligands in SSc skin lesions. Our study sheds light on the TLR4-IRF5 pathway in the pathology of SSc with clinical implications of targeting the IRF5 pathways in the suppression of disease development.


Assuntos
Fatores Reguladores de Interferon/fisiologia , Escleroderma Sistêmico/fisiopatologia , Receptor 4 Toll-Like/fisiologia , Animais , Linfócitos B/imunologia , Feminino , Fatores Reguladores de Interferon/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA