Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 153
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33563757

RESUMO

Sepsis is a major cause of mortality in intensive care units, which results from a severely dysregulated inflammatory response that ultimately leads to organ failure. While antibiotics can help in the early stages, effective strategies to curtail inflammation remain limited. The high mobility group (HMG) proteins are chromosomal proteins with important roles in regulating gene transcription. While HMGB1 has been shown to play a role in sepsis, the role of other family members including HMGXB4 remains unknown. We found that expression of HMGXB4 is strongly induced in response to lipopolysaccharide (LPS)-elicited inflammation in murine peritoneal macrophages. Genetic deletion of Hmgxb4 protected against LPS-induced lung injury and lethality and cecal ligation and puncture (CLP)-induced lethality in mice, and attenuated LPS-induced proinflammatory gene expression in cultured macrophages. By integrating genome-wide transcriptome profiling and a publicly available ChIP-seq dataset, we identified HMGXB4 as a transcriptional activator that regulates the expression of the proinflammatory gene, Nos2 (inducible nitric oxide synthase 2) by binding to its promoter region, leading to NOS2 induction and excessive NO production and tissue damage. Similar to Hmgxb4 ablation in mice, administration of a pharmacological inhibitor of NOS2 robustly decreased LPS-induced pulmonary vascular permeability and lethality in mice. Additionally, we identified the cell adhesion molecule, ICAM1, as a target of HMGXB4 in endothelial cells that facilitates inflammation by promoting monocyte attachment. In summary, our study reveals a critical role of HMGXB4 in exacerbating endotoxemia via transcriptional induction of Nos2 and Icam1 gene expression and thus targeting HMGXB4 may be an effective therapeutic strategy for the treatment of sepsis.


Assuntos
Endotoxemia/metabolismo , Animais , Células Endoteliais/metabolismo , Endotoxemia/etiologia , Endotoxemia/genética , Feminino , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Lipopolissacarídeos/toxicidade , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Transcriptoma
2.
Inflamm Res ; 72(8): 1539-1549, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37453943

RESUMO

BACKGROUND: Innate monocytes can adopt dynamic "memory" states ranging from low-grade inflammation to pathogenic exhaustion, dependent upon signal strength and history of challenges. Low-grade inflammatory monocytes facilitate the pathogenesis of chronic inflammatory diseases, while exhausted monocytes drive the pathogenesis of severe sepsis. Although clinical and basic studies suggest the conservation of key features of exhausted monocytes from human and murine sepsis, systems analyses of monocyte exhaustion among human and murine monocytes are lacking. METHODS: We performed cross examination of septic monocytes scRNAseq data recently collected from human sepsis patients as well as experimental septic mice, in reference to monocytes experimentally exhausted in vitro. Furthermore, we performed pseudo-time analyses of in vitro programmed monocytes following prolonged challenges causing either low-grade inflammation or exhaustion. Additional comparative analyses of low-grade inflammatory monocytes were performed with scRNAseq data from selected human patients with chronic low-grade inflammatory diseases. RESULTS: Our systems analyses reveal key features of monocyte exhaustion including reduced differentiation, pathogenic inflammation and immune suppression that are highly conserved in human and murine septic monocytes, and captured by in vitro experimental exhaustion. Pseudo-time analyses reveal that monocytes initially transition into a less-differentiated state with proliferative potential. The expansion of proliferative monocytes can be observed not only in experimentally challenged monocytes, but also in tissues of murine sepsis and human septic blood. We observed that monocytes similarly transition into the less-differentiated state when challenged with a subclinical dose endotoxin under chronic inflammatory conditions. Instead of being exhausted, monocytes with prolonged challenges with super-low dose endotoxin bifurcate into the low-grade inflammatory immune-enhancing or the chemotactic/adhesive state, often see in atherosclerosis or auto-immune diseases. CONCLUSIONS: Key features of monocyte memory dynamics are identified and conserved in human and murine monocytes, which can be captured by prolonged challenges of innate signals with varying signal strength.


Assuntos
Monócitos , Sepse , Humanos , Animais , Camundongos , Sepse/patologia , Inflamação/patologia , Endotoxinas
3.
Inflamm Res ; 72(8): 1733-1744, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37563334

RESUMO

OBJECTIVE: Proper inflammation resolution is crucial to prevent runaway inflammation during sepsis and reduce sepsis-related mortality/morbidity. Previous studies suggest that deleting TRAM, a key TLR4 signaling adaptor, can reprogram the first inflammatory responder cell-neutrophil from an inflammatory state to a resolving state. In this study, we aim to examine the therapeutic potential of TRAM-deficient neutrophils in vivo with recipient mice undergoing experimental sepsis. MATERIAL AND METHODS: Wild-type or Tram-/- mice were intraperitoneally injected with cecal slurry to induce either severe or mild sepsis. Phenotypic examinations of sepsis and neutrophil characteristics were examined in vivo and ex vivo. The propagations of resolution from donor neutrophils to recipient cells such as monocytes, T cells, and endothelial cells were examined through co-culture assays in vitro. The efficacies of Tram-/- neutrophils in reducing inflammation were studied by transfusing either wild-type or Tram-/- neutrophils into septic recipient mice. RESULTS: Tram-/- septic mice had improved survival and attenuated injuries within the lung and kidney tissues as compared to wild-type septic mice. Wild-type septic mice transfused with Tram-/- resolving neutrophils exhibited reduced multi-organ damages and improved cellular homeostasis. In vitro co-culture studies revealed that donor Tram-/- neutrophils can effectively propagate cellular homeostasis to co-cultured neighboring monocytes, neutrophils, T cells as well as endothelial cells. CONCLUSIONS: Neutrophils with TRAM deletion render effective reprogramming into a resolving state beneficial for ameliorating experimental sepsis, with therapeutic potential in propagating cellular and tissue homeostasis as well as treating sepsis.


Assuntos
Neutrófilos , Sepse , Animais , Camundongos , Células Endoteliais , Inflamação , Camundongos Endogâmicos C57BL , Sepse/genética , Sepse/terapia
4.
Inflamm Res ; 72(5): 1083-1097, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37060359

RESUMO

OBJECTIVE: Patients with systemic lupus erythematosus (SLE) often develop multi-organ damages including heart and kidney complications. We sought to better define the underlying mechanisms with a focus on the chemokine receptor CX3CR1. METHODS: We generated Cx3cr1-deficient MRL/lpr lupus-prone mice through backcrossing. We then employed heterozygous intercross to generate MRL/lpr littermates that were either sufficient or deficient of CX3CR1. The mice were also treated with either Lactobacillus spp. or a high-fat diet (HFD) followed by assessments of the kidney and heart, respectively. RESULTS: Cx3cr1-/- MRL/lpr mice exhibited a distinct phenotype of exacerbated glomerulonephritis compared to Cx3cr1+/+ littermates, which was associated with a decrease of spleen tolerogenic marginal zone macrophages and an increase of double-negative T cells. Interestingly, upon correction of the gut microbiota with Lactobacillus administration, the phenotype of exacerbated glomerulonephritis was reversed, suggesting that CX3CR1 controls glomerulonephritis in MRL/lpr mice through a gut microbiota-dependent mechanism. Upon treatment with HFD, Cx3cr1-/- MRL/lpr mice developed significantly more atherosclerotic plaques that were promoted by Ly6C+ monocytes. Activated monocytes expressed ICOS-L that interacted with ICOS-expressing follicular T-helper cells, which in turn facilitated a germinal center reaction to produce more autoantibodies. Through a positive feedback mechanism, the increased circulatory autoantibodies further promoted the activation of Ly6C+ monocytes and their display of ICOS-L. CONCLUSIONS: We uncovered novel, Cx3cr1 deficiency-mediated pathogenic mechanisms contributing to SLE-associated glomerulonephritis and cardiovascular disease.


Assuntos
Doenças Cardiovasculares , Glomerulonefrite , Lúpus Eritematoso Sistêmico , Animais , Camundongos , Receptor 1 de Quimiocina CX3C/genética , Camundongos Endogâmicos MRL lpr , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/genética , Autoanticorpos , Modelos Animais de Doenças
5.
J Immunol ; 206(12): 2980-2988, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34031144

RESUMO

Low-grade inflammatory monocytes critically contribute to the pathogenesis of chronic inflammatory diseases such as atherosclerosis. The elevated expression of coactivating molecule CD40 as well as key adhesion molecule CD11a is a critical signature of inflammatory monocytes from both human patients with coronary artery diseases as well as in animal models of atherosclerosis. In this study, we report that subclinical superlow-dose LPS, a key risk factor for low-grade inflammation and atherosclerosis, can potently trigger the induction of CD40 and CD11a on low-grade inflammatory monocytes. Subclinical endotoxin-derived monocytes demonstrate immune-enhancing effects and suppress the generation of regulatory CD8+CD122+ T cells, which further exacerbate the inflammatory environment conducive for chronic diseases. Mechanistically, subclinical endotoxemia activates TRAM-mediated signaling processes, leading to the activation of MAPK and STAT5, which is responsible for the expression of CD40 and CD11a. We also demonstrate that TRAM-mediated monocyte polarization can be suppressed by IRAK-M. IRAK-M-deficient monocytes have increased expression of TRAM, elevated induction of CD40 and CD11a by subclinical-dose endotoxin, and are more potent in suppressing the CD8 regulatory T cells. Mice with IRAK-M deficiency generate an increased population of inflammatory monocytes and a reduced population of CD8 T regulatory cells. In contrast, mice with TRAM deficiency exhibit a significantly reduced inflammatory monocyte population and an elevated CD8 T regulatory cell population. Together, our data reveal a competing intracellular circuitry involving TRAM and IRAK-M that modulate the polarization of low-grade inflammatory monocytes with an immune-enhancing function.


Assuntos
Moléculas de Adesão Celular/imunologia , Inflamação/imunologia , Quinases Associadas a Receptores de Interleucina-1/imunologia , Monócitos/imunologia , Receptores de Interleucina/imunologia , Receptor 4 Toll-Like/imunologia , Animais , Células Cultivadas , Quinases Associadas a Receptores de Interleucina-1/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
Immunity ; 39(2): 272-85, 2013 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-23973223

RESUMO

Regulatory T (Treg) cells suppress inflammatory immune responses and autoimmunity caused by self-reactive T cells. The key Treg cell transcription factor Foxp3 is downregulated during inflammation to allow for the acquisition of effector T cell-like functions. Here, we demonstrate that stress signals elicited by proinflammatory cytokines and lipopolysaccharides lead to the degradation of Foxp3 through the action of the E3 ubiquitin ligase Stub1. Stub1 interacted with Foxp3 to promote its K48-linked polyubiquitination in an Hsp70-dependent manner. Knockdown of endogenous Stub1 or Hsp70 prevented Foxp3 degradation. Furthermore, the overexpression of Stub1 in Treg cells abrogated their ability to suppress inflammatory immune responses in vitro and in vivo and conferred a T-helper-1-cell-like phenotype. Our results demonstrate the critical role of the stress-activated Stub1-Hsp70 complex in promoting Treg cell inactivation, thus providing a potential therapeutic target for the intervention against autoimmune disease, infection, and cancer.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Células Cultivadas , Citocinas/metabolismo , Inibidores Enzimáticos , Células HEK293 , Proteínas de Choque Térmico HSP70/genética , Humanos , Imidazóis , Inflamação/genética , Inflamação/imunologia , Lipopolissacarídeos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Fenótipo , Piridinas , Interferência de RNA , RNA Interferente Pequeno , Linfócitos T Auxiliares-Indutores/imunologia , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
7.
Handb Exp Pharmacol ; 276: 43-64, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34486096

RESUMO

Neutrophils, the most abundant leukocytes in circulation and the first responders to infection and inflammation, closely modulate both acute and chronic inflammatory processes. Resting neutrophils constantly patrol vasculature and migrate to tissues when challenges occur. When infection and/or inflammation recede, tissue neutrophils will be subsequently cleaned up by macrophages which collectively contribute to the resolution of inflammation. While most studies focus on the anti-microbial function of neutrophils including phagocytosis, degranulation, and neutrophil extracellular traps (NETs) formation, recent research highlighted additional contributions of neutrophils beyond simply controlling infectious agents. Neutrophils with resolving characteristics may alter the activities of neighboring cells and facilitate inflammation resolution, modulate long-term macrophage and adaptive immune responses, therefore having important impacts on host pathophysiology. The focus of this chapter is to provide an updated assessment of recent progress in the emerging field of neutrophil programming and memory in the context of both acute and chronic diseases.


Assuntos
Armadilhas Extracelulares , Neutrófilos , Humanos , Imunidade Inata , Inflamação , Macrófagos , Neutrófilos/patologia , Fagocitose
8.
Handb Exp Pharmacol ; 276: 23-41, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34085119

RESUMO

Innate immunity exhibits memory characteristics, reflected not only in selective recognition of external microbial or internal damage signals, but more importantly in history and signal-strength dependent reprogramming of innate leukocytes characterized by priming, tolerance, and exhaustion. Key innate immune cells such as monocytes and neutrophils can finely discern and attune to the duration and intensity of external signals through rewiring of internal signaling circuitries, giving rise to a vast array of discreet memory phenotypes critically relevant to managing tissue homeostasis as well as diverse repertoires of inflammatory conditions. This review will highlight recent advances in this rapidly expanding field of innate immune programming and memory, as well as its translational implication in the pathophysiology of selected inflammatory diseases.


Assuntos
Imunidade Inata , Memória Imunológica , Humanos , Tolerância Imunológica , Monócitos , Transdução de Sinais
9.
Mol Ther ; 28(1): 89-99, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31607540

RESUMO

Tumor-associated immune-suppressive neutrophils are prevalent in various cancers, including colorectal cancer. However, mechanisms of immune-suppressive neutrophils are not well understood. We report that a key innate suppressor, IRAK-M (interleukin-1 receptor-associated kinase M), is critically involved in the establishment of immune-suppressive neutrophils. In contrast to the wild-type (WT) neutrophils exhibiting immune-suppressive signatures of CD11bhighPD-L1highCD80low, IRAK-M-deficient neutrophils are rewired with reduced levels of inhibitory molecules PD-L1 and CD11b, as well as enhanced expression of stimulatory molecules CD80 and CD40. The reprogramming of IRAK-M-deficient neutrophils is mediated by reduced activation of STAT1/3 and enhanced activation of STAT5. As a consequence, IRAK-M-deficient neutrophils demonstrate enhanced capability to promote, instead of suppress, the proliferation and activation of effector T cells both in vitro and in vivo. Functionally, we observed that the transfusion of IRAK-M-/- neutrophils can potently render an enhanced anti-tumor immune response in the murine inflammation-induced colorectal cancer model. Collectively, our study defines IRAK-M as an innate suppressor for neutrophil function and reveals IRAK-M as a promising target for rewiring neutrophils in anti-cancer immunotherapy.


Assuntos
Transferência Adotiva/métodos , Neoplasias Colorretais/terapia , Imunidade Inata/genética , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Neutrófilos/imunologia , Animais , Azoximetano/farmacologia , Antígeno B7-1/metabolismo , Antígeno B7-H1/metabolismo , Antígenos CD11/metabolismo , Antígenos CD40/metabolismo , Colite/induzido quimicamente , Colite/complicações , Neoplasias Colorretais/etiologia , Modelos Animais de Doenças , Inflamação/induzido quimicamente , Inflamação/metabolismo , Quinases Associadas a Receptores de Interleucina-1/genética , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , Neutrófilos/metabolismo , Linfócitos T/imunologia , Resultado do Tratamento
10.
J Biol Chem ; 293(31): 12239-12247, 2018 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-29921584

RESUMO

Toll-interacting protein (Tollip) deficiency has been implicated in complex inflammatory and infectious diseases whose mechanisms are poorly understood. Comparing the gene expression profiles of WT and Tollip-deficient murine embryonic fibroblasts, we observed here that Tollip deficiency selectively reduces the expression of the inflammatory cytokines interleukin 6 (IL-6), IL-12, and tumor necrosis factor α (TNFα) but potentiates the expression of fatty acid-binding protein 4 (FABP4) in these cells. We also observed that expression of hypoxia-inducible factor 1-α (HIF1α) is reduced, whereas that of signal transducer and activator of transcription 5 (STAT5) is elevated, in Tollip-deficient cells, correlating with the decreased expression of inflammatory cytokines and increased expression of FABP4 in these cells. We further found that the coupling of ubiquitin to ER degradation (CUE) domain of Tollip is required for stimulating HIF1α activity, because Tollip CUE-domain mutant cells exhibited reduced levels of HIF1α and selected cytokines. Tollip is known to mediate autophagy and lysosome fusion, and herein we observed that Tollip's autophagy function is required for modulating STAT5 and FABP4 expression. Bafilomycin A, an inhibitor of lysosome fusion, enhanced STAT5 and FABP4 expression in WT fibroblasts, whereas torin 2, an activator of autophagy, reduced STAT5 and FABP4 expression in Tollip-deficient fibroblasts. Taken together, our study reveals that Tollip differentially modulates HIF1α and STAT5 expression in fibroblasts, potentially explaining the complex and context-dependent contribution of Tollip to disease development.


Assuntos
Fibroblastos/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fator de Transcrição STAT5/genética , Animais , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Interleucina-12/genética , Interleucina-12/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Knockout , Ligação Proteica , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais
11.
J Autoimmun ; 102: 50-64, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31080014

RESUMO

Accumulating evidence suggests granulocyte macrophage-colony stimulating factor (GM-CSF) can function as an inflammatory mediator, but whether GM-CSF-producing CD4+ T cells (TH-GM-CSF) are a distinct T helper cell subset is lacking. Herein we demonstrate that interleukin (IL)-1ß exclusively drives differentiation of naïve CD4+ T cells into TH-GM-CSF cells via inducing ubiquitination of IL-1 receptor-associated kinase 1 (IRAK1) and subsequent activation of the transcription factor NF-kappaB (NF-κB), independent of RAR-related orphan receptor gamma (RORγt) required for TH17 differentiation. In vivo, TH-GM-CSF cells are present in murine Citrobacter Rodentium infections and mediate colitis following adoptive transfer of CD4+ T cells into Rag1-/- mice via GM-CSF-induced macrophage activation. The TH-GM-CSF cell phenotype is stable and distinct from the TH17 genetic program, but IL-1ß can convert pre-formed TH17 cells into TH-GM-CSF cells, thereby accounting for previously reported associations between IL-17 and GM-CSF. Together, our results newly identify IL-1ß/NF-κB-dependent TH-GM-CSF cells as a unique T helper cell subset and highlight the importance of CD4+ T cell-derived GM-CSF induced macrophage activation as a previously undescribed T cell effector mechanism.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Interleucina-1beta/imunologia , Ativação de Macrófagos/imunologia , Células Th17/citologia , Células Th17/imunologia , Animais , Diferenciação Celular/imunologia , Citrobacter rodentium/imunologia , Colite/imunologia , Inflamação/imunologia , Inflamação/patologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Células Th17/patologia , Ubiquitinação
12.
J Autoimmun ; 96: 113-122, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30241692

RESUMO

The follicular helper T cell (TFH) are established regulators of germinal center (GC) B cells, whether TFH have pathogenic potential independent of B cells is unknown. Based on in vitro TFH cell differentiation, in vivo T cell transfer animal colitis model, and intestinal tissues of inflammatory bowel disease (IBD) patients, TFH and its functions in colitis development were analyzed by FACS, ChIP, ChIP-sequencing, WB, ELISA and PCR. Herein we demonstrate that intestinal tissues of patients and colon tissues obtained from Rag1-/- recipients of naïve CD4+ T cells with colitis, each over-express TFH-associated gene products. Adoptive transfer of naïve Bcl6-/- CD4+ T cells into Rag1-/- recipient mice abrogated development of colitis and limited TFH differentiation in vivo, demonstrating a mechanistic link. In contrast, T cell deficiency of interferon regulatory factor 8 (IRF8) resulted in augmentation of TFH induction in vitro and in vivo. Functional studies showed that adoptive transfer of IRF8 deficient CD4+ T cells into Rag1-/- recipients exacerbated colitis development associated with increased gut TFH-related gene expression, while Irf8-/-/Bcl6-/- CD4+ T cells abrogated colitis, together indicating that IRF8-regulated TFH can directly cause colon inflammation. Molecular analyses revealed that IRF8 suppresses TFH differentiation by inhibiting transcription and transactivation of the TF IRF4, which is also known to be essential for TFH induction. Our documentation showed that IRF8-regulated TFH can function as B-cell-independent, pathogenic, mediators of colitis suggests that targeting TFH could be effective for treatment of IBD.


Assuntos
Linfócitos B/imunologia , Colite/imunologia , Colo/metabolismo , Doença de Crohn/imunologia , Centro Germinativo/imunologia , Fatores Reguladores de Interferon/metabolismo , Linfócitos T Auxiliares-Indutores/imunologia , Transferência Adotiva , Animais , Células Cultivadas , Colite/genética , Colo/patologia , Doença de Crohn/genética , Modelos Animais de Doenças , Humanos , Fatores Reguladores de Interferon/genética , Ativação Linfocitária , Camundongos , Camundongos Knockout , Comunicação Parácrina , Proteínas Proto-Oncogênicas c-bcl-6/genética , Linfócitos T Auxiliares-Indutores/transplante
13.
Bull Math Biol ; 81(1): 256-276, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30387078

RESUMO

The size of primary challenge with lipopolysaccharide induces changes in the innate immune cells phenotype between pro-inflammatory and pro-tolerant states when facing a secondary lipopolysaccharide challenge. To determine the molecular mechanisms governing this differential response, we propose a mathematical model for the interaction between three proteins involved in the immune cell decision making: IRAK-1, PI3K, and RelB. The mutual inhibition of IRAK-1 and PI3K in the model leads to bistable dynamics. By using the levels of RelB as indicative of strength of the immune responses, we connect the size of different primary lipopolysaccharide doses to the differential phenotypical outcomes following a secondary challenge. We further predict under what circumstances the primary LPS dose does not influence the response to a secondary challenge. Our results can be used to guide treatments for patients with either autoimmune disease or compromised immune system.


Assuntos
Imunidade Inata , Modelos Imunológicos , Animais , Simulação por Computador , Humanos , Tolerância Imunológica , Quinases Associadas a Receptores de Interleucina-1/imunologia , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Conceitos Matemáticos , Fenótipo , Fosfatidilinositol 3-Quinases/imunologia , Fator de Transcrição RelB/imunologia
14.
J Biol Chem ; 292(29): 12339-12350, 2017 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-28572512

RESUMO

Chronic inflammation may contribute to insulin resistance via molecular cross-talk between pathways for pro-inflammatory and insulin signaling. Interleukin 1 receptor-associated kinase 1 (IRAK-1) mediates pro-inflammatory signaling via IL-1 receptor/Toll-like receptors, which may contribute to insulin resistance, but this hypothesis is untested. Here, we used male Irak1 null (k/o) mice to investigate the metabolic role of IRAK-1. C57BL/6 wild-type (WT) and k/o mice had comparable body weights on low-fat and high-fat diets (LFD and HFD, respectively). After 12 weeks on LFD (but not HFD), k/o mice (versus WT) had substantially improved glucose tolerance (assessed by the intraperitoneal glucose tolerance test (IPGTT)). As assessed with the hyperinsulinemic euglycemic glucose clamp technique, insulin sensitivity was 30% higher in the Irak1 k/o mice on chow diet, but the Irak1 deletion did not affect IPGTT outcomes in mice on HFD, suggesting that the deletion did not overcome the impact of obesity on glucose tolerance. Moreover, insulin-stimulated glucose-disposal rates were higher in the k/o mice, but we detected no significant difference in hepatic glucose production rates (± insulin infusion). Positron emission/computed tomography scans indicated higher insulin-stimulated glucose uptake in muscle, but not liver, in Irak1 k/o mice in vivo Moreover, insulin-stimulated phosphorylation of Akt was higher in muscle, but not in liver, from Irak1 k/o mice ex vivo In conclusion, Irak1 deletion improved muscle insulin sensitivity, with the effect being most apparent in LFD mice.


Assuntos
Intolerância à Glucose/metabolismo , Resistência à Insulina , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Músculo Esquelético/metabolismo , Animais , Biomarcadores/sangue , Biomarcadores/metabolismo , Cruzamentos Genéticos , Dieta com Restrição de Gorduras , Dieta Hiperlipídica/efeitos adversos , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Técnica Clamp de Glucose , Intolerância à Glucose/etiologia , Intolerância à Glucose/fisiopatologia , Intolerância à Glucose/prevenção & controle , Hemizigoto , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Quinases Associadas a Receptores de Interleucina-1/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/enzimologia , Obesidade/etiologia , Obesidade/fisiopatologia , Especificidade de Órgãos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Gordura Subcutânea Abdominal/efeitos dos fármacos , Gordura Subcutânea Abdominal/enzimologia , Gordura Subcutânea Abdominal/metabolismo
15.
Biochem Biophys Res Commun ; 501(2): 563-569, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29750958

RESUMO

IL-22 plays an important role in tissue repair and inflammatory responses, and is implicated in the pathogenesis of psoriasis, ulcerative colitis, as well as liver and pancreas damage. The molecular mechanisms of its regulation have been actively studied. Here, we show that the differential regulation of IL-22 expression in CD4+ T cells by IL-6 and IL-27 was detected rapidly after stimulation. Chromatin immunoprecipitation (ChIP) and luciferase reporter assays demonstrated that both STAT1 and STAT3 directly bind to the STAT responsive elements (SRE) of the IL-22 promoter, and the balance between activated STAT3 and STAT1 determines IL-22 promoter activities. We further show that the heterozygous mutation of the STAT1 gene results in elevated levels of IL-22 production and induces much severer skin inflammation in an imiquimod (IMQ)-induced murine psoriasis model. Together, our results reveal a novel regulatory mechanism of IL-22 expression by STAT1 through directly antagonizing STAT3, and the importance of the balance between STAT3 and STAT1 in IL-22 regulation and psoriasis pathogenesis.


Assuntos
Regulação da Expressão Gênica , Interleucinas/genética , Psoríase/genética , Fator de Transcrição STAT1/genética , Pele/patologia , Animais , Interleucinas/imunologia , Camundongos Endogâmicos C57BL , Mutação , Regiões Promotoras Genéticas , Psoríase/imunologia , Psoríase/patologia , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT3/imunologia , Pele/imunologia , Interleucina 22
16.
J Immunol ; 196(5): 2300-2308, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26810228

RESUMO

Subclinical circulating bacterial endotoxin LPS has been implicated as an important cofactor in the development and progression of nonalcoholic steatohepatitis, but the underlying mechanisms remain unclear. In this study, we demonstrated that 4-wk injection with superlow-dose LPS significantly promoted neutrophil infiltration and accelerated nonalcoholic steatohepatitis progression, including exacerbated macrovesicular steatosis, inflammation, and hepatocyte ballooning in high-fat diet-fed apolipoprotein E knockout mice. This effect could sustain for a month after stoppage of LPS injection. LPS also significantly increased numbers of apoptotic nuclei in hepatocytes and expressions of proapoptotic regulators. Moreover, LPS sustained the low-grade activation of p38 MAPK and inhibited the expression of the upstream MAPK phosphatase 7. By applying selective inhibitors, we demonstrated that the activation of p38 MAPKs is required for neutrophil migration induced by superlow-dose LPS in vitro. Together, these data suggest that superlow-dose LPS may sustain the low-grade activation of p38 MAPKs and neutrophil infiltration, leading to the exacerbation of steatohepatitis.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Endotoxinas/administração & dosagem , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/patologia , Animais , Apolipoproteínas E/deficiência , Apoptose/imunologia , Quimiotaxia de Leucócito , Modelos Animais de Doenças , Progressão da Doença , Fosfatases de Especificidade Dupla/metabolismo , Metabolismo dos Lipídeos , Lipídeos/sangue , Lipopolissacarídeos/administração & dosagem , Fígado/metabolismo , Sistema de Sinalização das MAP Quinases , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Modelos Biológicos , Infiltração de Neutrófilos/imunologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Fosforilação , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2796-2807, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28802852

RESUMO

Autophagy extensively participates in immune responses and inflammatory diseases. Myeloid-derived suppressor cells (MDSCs) are derived from CD11b+Gr1+ cells under pathological conditions and play an immunosuppressive role in the pathogenesis of cancer and inflammatory diseases. However, the role of autophagy in regulating the accumulation and activity of MDSCs remains unknown. In the present study, we evaluated the effects and mechanisms of autophagy on regulating accumulation and activity of MDSCs. We first found that granulocytic MDSCs (G-MDSCs), but not monocytic MDSCs (M-MDSCs), were accumulated in mice challenged by lipopolysaccharide (LPS) and showed an elevated autophagy activity. Pharmacological inhibition of autophagy significantly enhanced accumulation of G-MDSCs in vivo and in vitro. Notably, inhibition of autophagy enhanced the immunosuppressive activity of G-MDSCs on M1 macrophage polarization by promoting reactive oxygen species (ROS) production. Inhibition of autophagy promotes the phosphorylation of signal transducer and activator of transcription 3 (STAT3) in G-MDSCs, which is required for the accumulation and activity of MDSCs. In addition, in vivo pharmacological inhibition of autophagy significantly attenuated the condition of mice challenged by LPS. Thus, we conclude that inhibition of autophagy contributes to accumulation and immunosuppressive function of G-MDSCs by promoting the activation of STAT3 signaling, suggesting that autophagy may play a critical role in regulating accumulation and activity of MDSCs. Our study provides new insights into understanding the mechanisms of autophagy in regulating immune responses and pathogenesis of inflammatory diseases.


Assuntos
Autofagia/imunologia , Granulócitos/imunologia , Células Supressoras Mieloides/imunologia , Fator de Transcrição STAT3/imunologia , Choque Séptico/imunologia , Transdução de Sinais/imunologia , Animais , Autofagia/efeitos dos fármacos , Granulócitos/patologia , Lipopolissacarídeos/toxicidade , Camundongos , Células Supressoras Mieloides/patologia , Choque Séptico/induzido quimicamente , Choque Séptico/patologia , Transdução de Sinais/efeitos dos fármacos
18.
Brain Behav Immun ; 59: 200-210, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27720815

RESUMO

The excessive accumulation of specific cellular proteins or autophagic vacuoles (AVs) within neurons is a pathologic hallmark of neurodegenerative diseases. Constitutive autophagy in neurons prevents abnormal intracellular protein aggregation and is critical for maintaining cell survival. Since our previous study showed that Toll-interacting protein (Tollip)-deficient macrophages had constitutive disruption of endosome-lysosome fusion, we hypothesize that Tollip deficiency may also promote neuron death via blockage of autophagy completion. Indeed, we observed significantly higher levels of neuron death in the brain regions of cerebral cortex, hippocampus, and cerebellum from ApoE-/-/Tollip-/- mice as compared to ApoE-/- mice fed with high fat diet (HFD). We further documented diminished density of neurons and increased ratios of TUNEL positive cells in the hippocampus of ApoE-/-/Tollip-/- mice. The ultrastructural electron microscopy analyses revealed neuron cell shrinkage as well as loss of intracellular structure in brain tissues from ApoE-/-/Tollip-/- mice. There was dramatic accumulation of autophagosomes in the cytoplasm, elevated accumulation of ß-amyloid and α-synuclein, and increased levels of p62 and Parkin in the brain tissues from ApoE-/-/Tollip-/- mice as compared to ApoE-/- mice. Our data suggest that Tollip may play a crucial role in sustaining neuron health by facilitating the completion of autophagy, and that Tollip-deficiency may accelerate neuron death related to neurological disease such as Alzheimer's disease.


Assuntos
Apolipoproteínas E/deficiência , Autofagia/genética , Dieta Hiperlipídica , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Degeneração Neural/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Encéfalo/citologia , Região CA1 Hipocampal/patologia , Tamanho Celular , Endossomos/fisiologia , Lisossomos/fisiologia , Macrófagos/fisiologia , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Fagossomos/fisiologia , alfa-Sinucleína/metabolismo
19.
J Pathol ; 238(4): 571-83, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26690561

RESUMO

Impaired wound healing often accompanies low-grade inflammatory conditions, during which circulating levels of subclinical super-low-dose endotoxin may persist. Low-grade inflammatory monocyte polarization may occur during chronic inflammation and deter effective wound repair. However, little is understood about the potential mechanisms of monocyte polarization by sustained insult of subclinical super-low-dose endotoxin. We observed that super-low-dose endotoxin preferentially programmes a low-grade inflammatory monocyte state in vitro and in vivo, as represented by the elevated population of CD11b(+) Ly6C(high) monocytes and sustained expression of CCR5. Mechanistically, super-low-dose endotoxin caused cellular stress, altered lysosome function and increased the transcription factor IRF5. TUDCA, a potent inhibitor of cellular stress, effectively blocked monocyte polarization and improved wound healing in mice injected with super-low-dose endotoxin. Our data revealed the polarization of low-grade inflammatory monocytes by sustained endotoxin challenge, its underlying mechanisms and a potential intervention strategy. Copyright © 2015 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Polaridade Celular/imunologia , Inflamação/patologia , Lisossomos/metabolismo , Monócitos/citologia , Cicatrização/fisiologia , Humanos , Macrófagos/citologia , Macrófagos/imunologia , Fatores de Transcrição/metabolismo , Cicatrização/imunologia
20.
Exp Appl Acarol ; 73(3-4): 429-438, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29197022

RESUMO

Coxiella-like endosymbiont (CLS-Hl) is a primary endosymbiont of Haemaphysalis longicornis. CLS-Hl infects tick special tissues and its prevalence is 100% in ovaries and Malpighian tubules. Tetracycline was injected into females, which then fed on rabbits also treated with tetracycline. The densities of CLS-Hl were measured by semi-quantitative PCR. CLS-Hl densities in ovaries and Malpighian tubes of H. longicornis had significant effects on engorged weight, feeding time, number of eggs, oviposition period, and hatching period. These findings suggested that CLS-Hl plays a role in the reproduction and development of H. longicornis.


Assuntos
Coxiella/fisiologia , Ixodidae/microbiologia , Ixodidae/fisiologia , Oviposição , Simbiose , Animais , Comportamento Alimentar , Feminino , Ixodidae/crescimento & desenvolvimento , Túbulos de Malpighi/microbiologia , Ovário/microbiologia , Reprodução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA