Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
EMBO J ; 41(4): e106825, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35023164

RESUMO

Despite extensive analysis of pRB phosphorylation in vitro, how this modification influences development and homeostasis in vivo is unclear. Here, we show that homozygous Rb∆K4 and Rb∆K7 knock-in mice, in which either four or all seven phosphorylation sites in the C-terminal region of pRb, respectively, have been abolished by Ser/Thr-to-Ala substitutions, undergo normal embryogenesis and early development, notwithstanding suppressed phosphorylation of additional upstream sites. Whereas Rb∆K4 mice exhibit telomere attrition but no other abnormalities, Rb∆K7 mice are smaller and display additional hallmarks of premature aging including infertility, kyphosis, and diabetes, indicating an accumulative effect of blocking pRb phosphorylation. Diabetes in Rb∆K7 mice is insulin-sensitive and associated with failure of quiescent pancreatic ß-cells to re-enter the cell cycle in response to mitogens, resulting in induction of DNA damage response (DDR), senescence-associated secretory phenotype (SASP), and reduced pancreatic islet mass and circulating insulin level. Pre-treatment with the epigenetic regulator vitamin C reduces DDR, increases cell cycle re-entry, improves islet morphology, and attenuates diabetes. These results have direct implications for cell cycle regulation, CDK-inhibitor therapeutics, diabetes, and longevity.


Assuntos
Envelhecimento/fisiologia , Ácido Ascórbico/farmacologia , Diabetes Mellitus Experimental/prevenção & controle , Proteína do Retinoblastoma/metabolismo , Animais , Senescência Celular/efeitos dos fármacos , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Fator de Transcrição E2F1/metabolismo , Desenvolvimento Embrionário/genética , Feminino , Fibroblastos/efeitos dos fármacos , Técnicas de Introdução de Genes , Células Secretoras de Insulina/patologia , Camundongos , Fosforilação , Gravidez , Proteína do Retinoblastoma/genética , Telômero/genética
2.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34301890

RESUMO

Cytosolic lipopolysaccharides (LPSs) bind directly to caspase-4/5/11 through their lipid A moiety, inducing inflammatory caspase oligomerization and activation, which is identified as the noncanonical inflammasome pathway. Galectins, ß-galactoside-binding proteins, bind to various gram-negative bacterial LPS, which display ß-galactoside-containing polysaccharide chains. Galectins are mainly present intracellularly, but their interactions with cytosolic microbial glycans have not been investigated. We report that in cell-free systems, galectin-3 augments the LPS-induced assembly of caspase-4/11 oligomers, leading to increased caspase-4/11 activation. Its carboxyl-terminal carbohydrate-recognition domain is essential for this effect, and its N-terminal domain, which contributes to the self-association property of the protein, is also critical, suggesting that this promoting effect is dependent on the functional multivalency of galectin-3. Moreover, galectin-3 enhances intracellular LPS-induced caspase-4/11 oligomerization and activation, as well as gasdermin D cleavage in human embryonic kidney (HEK) 293T cells, and it additionally promotes interleukin-1ß production and pyroptotic death in macrophages. Galectin-3 also promotes caspase-11 activation and gasdermin D cleavage in macrophages treated with outer membrane vesicles, which are known to be taken up by cells and release LPSs into the cytosol. Coimmunoprecipitation confirmed that galectin-3 associates with caspase-11 after intracellular delivery of LPSs. Immunofluorescence staining revealed colocalization of LPSs, galectin-3, and caspase-11 independent of host N-glycans. Thus, we conclude that galectin-3 amplifies caspase-4/11 oligomerization and activation through LPS glycan binding, resulting in more intense pyroptosis-a critical mechanism of host resistance against bacterial infection that may provide opportunities for new therapeutic interventions.


Assuntos
Caspases/metabolismo , Galectina 3/metabolismo , Inflamassomos/imunologia , Inflamação/imunologia , Lipopolissacarídeos/metabolismo , Macrófagos/imunologia , Animais , Citosol/metabolismo , Galectina 3/genética , Inflamassomos/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Piroptose
3.
Int J Mol Sci ; 23(3)2022 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-35163542

RESUMO

The PD-1/PD-L1 pathway is critical in T cell biology; however, the role of the PD-1/PD-L1 pathway in clinical characteristics and treatment outcomes in pulmonary tuberculosis (PTB) patients is unclear. We prospectively enrolled PTB, latent TB infection (LTBI), and non-TB, non-LTBI subjects. The expression of PD-1/PD-L1 on peripheral blood mononuclear cells (PBMCs) was measured and correlated with clinical characteristics and treatment outcomes in PTB patients. Immunohistochemistry and immunofluorescence were used to visualize PD-1/PD-L1-expressing cells in lung tissues from PTB patients and from murine with heat-killed MTB (HK-MTB) treatment. A total of 76 PTB, 40 LTBI, and 28 non-TB, non-LTBI subjects were enrolled. The expression of PD-1 on CD4+ T cells and PD-L1 on CD14+ monocytes was significantly higher in PTB cases than non-TB subjects. PTB patients with sputum smear/culture unconversion displayed higher PD-L1 expression on monocytes. PD-L1-expressing macrophages were identified in lung tissue from PTB patients, and co-localized with macrophages in murine lung tissues. Mycobacterium tuberculosis (MTB) whole cell lysate/EsxA stimulation of human and mouse macrophages demonstrated increased PD-L1 expression. In conclusion, increased expression of PD-L1 on monocytes in PTB patients correlated with higher bacterial burden and worse treatment outcomes. The findings suggest the involvement of the PD-1/PD-L1 pathway in MTB-related immune responses.


Assuntos
Antituberculosos/farmacologia , Antígeno B7-H1/metabolismo , Tuberculose Latente/metabolismo , Leucócitos Mononucleares/metabolismo , Mycobacterium tuberculosis/patogenicidade , Receptor de Morte Celular Programada 1/metabolismo , Tuberculose Pulmonar/metabolismo , Regulação para Cima , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Antituberculosos/uso terapêutico , Estudos de Casos e Controles , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Tuberculose Latente/microbiologia , Masculino , Camundongos , Pessoa de Meia-Idade , Mycobacterium tuberculosis/efeitos dos fármacos , Células THP-1 , Resultado do Tratamento , Tuberculose Pulmonar/tratamento farmacológico , Tuberculose Pulmonar/microbiologia , Adulto Jovem
4.
J Mol Cell Cardiol ; 155: 99-110, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33713645

RESUMO

Background Diabetes has a pronounced effect on the peripheral vasculature. The accumulation of advanced glycation end products (AGEs) is regarded as the crucial mechanism responsible for vascular damage in diabetes, but it is not easy to be avoided from food. In this study, we aimed to investigate the effects of an oral absorbent, AST-120, on the accumulation of AGEs and changes in blood flow recovery in diabetic mice. Methods The mice were divided into four groups, wild-type (WT) mice without treatment, WT mice treated with 5% AST-120 mixed into pulverized chow, streptozotocin-induced diabetes mellitus (DM) mice, and DM mice treated with 5% AST-120. Six weeks after hind-limb ischemia surgery, blood flow reperfusion, histology, plasma AGE, and cytokine were examined. Bone marrow cells were cultured and derived into macrophages to evaluate the effects of AGEs on macrophage polarization. Results Plasma AGEs were significantly increased in diabetic mice. AST-120 could bind to AGEs and reduced their plasma concentrations. Histological analysis revealed fewer collateral vessels with corresponding impairment of blood flow recovery in diabetic mice. In these mice, AGE-positive and AGE receptor-positive macrophages were numerous in ischemic limbs compared with non- diabetic mice. In diabetic mice, macrophages in ischemic tissues demonstrated greater M1 polarization than M2 polarization; this pattern was reversed in the AST-120 treatment group. The change in macrophage polarization was associated with the corresponding expression of pro-inflammatory cytokines in the ischemic tissues. In cell cultures, AGEs triggered the transformation of bone marrow-derived macrophages into the M1 phenotype. The alterations in the polarization of macrophages were reversed after treatment with AST-120. Conclusions Oral administration of AST-120 decreased the serum levels of AGEs in diabetic mice and improved neovascularization of ischemic limbs. This benefit may be due to, at least partially, the alterations in macrophage polarization and the associated changes in inflammatory cytokines.


Assuntos
Carbono/farmacologia , Plasticidade Celular/efeitos dos fármacos , Isquemia/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Músculos/irrigação sanguínea , Músculos/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Óxidos/farmacologia , Animais , Linhagem Celular , Citocinas/sangue , Citocinas/metabolismo , Diabetes Mellitus Experimental , Produtos Finais de Glicação Avançada/sangue , Produtos Finais de Glicação Avançada/metabolismo , Mediadores da Inflamação/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Modelos Biológicos , Músculos/efeitos dos fármacos
5.
Stem Cells ; 37(5): 631-639, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30681755

RESUMO

Induced pluripotent stem cells (iPSCs) can attenuate the pathological severity and neutrophil migration of lipopolysaccharide (LPS)-induced acute lung injury (ALI). However, interactions that may occur between iPSCs and the triggering receptor expressed on myeloid cells (TREM) family of proteins remain unclear. In this study, murine iPSCs (miPSCs) were delivered via tail vein injection to wild type, TREM-1 knockout (KO), and TREM-2 KO C57BL/6 mice 4 hours after an intratracheal delivery of LPS. Twenty-four hours later, the bronchoalveolar lavage fluid and lung tissue were collected to perform histology, immunohistochemistry, neutrophil counts, Western blot assays, and enzyme-linked immunosorbent assays. Neutrophils were also isolated from the bone marrow to perform in vitro migration assays. In the lung tissues collected, LPS increased the expression of TREM-1 and TREM-2, with the TREM-2 KO mice expressing more TREM-1 than the wild-type mice. The TREM-2 KO mice also exhibited greater severity of LPS-induced ALI, enhanced neutrophil infiltration in the lung tissues, and a higher ratio of phosphorylated p38 to total p38 (p-p38/p38) in neutrophils. The p-p38/p38 ratio and the expression of vascular cell adhesion molecule-1 and certain proinflammatory cytokines (macrophage inflammatory protein-2, tumor necrosis factor-α, interleukin-6, and interleukin-1ß) were increased in whole lung extracts following LPS-induced ALI, and these levels were even more in LPS-treated TREM-2 KO mice. These effects were reduced when miPSCs were administered. Thus, the results of this study suggest that miPSCs attenuate the role of neutrophils in lung inflammation and injury induced by LPS by reducing their expression of TREM-1 and p38 mitogen-activated protein kinase signaling. Stem Cells 2019;37:631-639.


Assuntos
Lesão Pulmonar Aguda/terapia , Células-Tronco Pluripotentes Induzidas/transplante , Infiltração de Neutrófilos/genética , Receptor Gatilho 1 Expresso em Células Mieloides/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Animais , Quimiocina CXCL2/genética , Endotoxinas/toxicidade , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Interleucina-1beta/genética , Lipopolissacarídeos/toxicidade , Pulmão/metabolismo , Camundongos , Camundongos Knockout , Células Mieloides/metabolismo , Fosforilação , Receptores Imunológicos/genética , Transdução de Sinais/genética
6.
Int J Mol Sci ; 21(17)2020 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-32825118

RESUMO

Mounting evidence indicates that an increase in histone deacetylation contributes to renal fibrosis. Although inhibition of histone deacetylase (HDAC) can reduce the extent of fibrosis, whether HDAC inhibitors exert the antifibrotic effect through modulating the phenotypes of macrophages, the key regulator of renal fibrosis, remains unknown. Moreover, the functional roles of the M2 macrophage subpopulation in fibrotic kidney diseases remain incompletely understood. Herein, we investigated the role of HDAC inhibitors on renal fibrogenesis and macrophage plasticity. We found that HDAC inhibition by trichostatin A (TSA) reduced the accumulation of interstitial macrophages, suppressed the activation of myofibroblasts and attenuated the extent of fibrosis in obstructive nephropathy. Moreover, TSA inhibited M1 macrophages and augmented M2 macrophage infiltration in fibrotic kidney tissue. Interestingly, TSA preferentially upregulated M2c macrophages and suppressed M2a macrophages in the obstructed kidneys, which was correlated with a reduction of interstitial fibrosis. TSA also repressed the expression of proinflammatory and profibrotic molecules in cultured M2a macrophages and inhibited the activation of renal myofibroblasts. In conclusion, our study was the first to show that HDAC inhibition by TSA alleviates renal fibrosis in obstructed kidneys through facilitating an M1 to M2c macrophage transition.


Assuntos
Inibidores de Histona Desacetilases/uso terapêutico , Ácidos Hidroxâmicos/uso terapêutico , Macrófagos/efeitos dos fármacos , Insuficiência Renal Crônica/tratamento farmacológico , Animais , Linhagem Celular , Células Cultivadas , Fibrose , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/metabolismo , Ratos , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia
7.
Int J Mol Sci ; 21(13)2020 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-32630825

RESUMO

Neutrophils are involved in the alveolitis of idiopathic pulmonary fibrosis (IPF). However, their pathogenic mechanisms are still poorly understood. Nintedanib has antifibrotic and anti-inflammatory activity in IPF. This study aimed to investigate the regulatory mechanism of nintedanib on neutrophil chemotaxis in bleomycin (BLM)-induced pulmonary fibrosis. Nintedanib was administered via oral gavage to male C57BL/6 mice 24 h after a bleomycin intratracheal injection (1.5 U/kg). Lung histopathological findings, the expression of cytokines, and the regulatory signaling pathways of neutrophil chemotaxis were analyzed. The effect of nintedanib was also investigated in a mouse model with adoptive neutrophil transfer in vivo. Nintedanib significantly decreased the histopathological changes and neutrophil recruitment in BLM-induced pulmonary fibrosis. Nintedanib mediated a downregulation of chemokine (C-X-C motif) receptor 2 (CXCR2) and very late antigen 4 (VLA-4) expression, as well as an upregulation of G protein-coupled receptor kinase 2 (GRK2) activity in peripheral blood neutrophils in BLM-induced pulmonary fibrosis. Nintedanib also decreased the activation of endothelial cells by the decreased expression of vascular cell adhesion molecule 1 (VCAM-1). The effect of nintedanib on regulating neutrophil chemotaxis was also confirmed by a mouse model with adoptive neutrophil transfer in vivo. In conclusion, nintedanib reduces neutrophil chemotaxis and endothelial cell activation to regulate the severity of BLM-induced pulmonary fibrosis. These effects are associated with an enhancement of GRK2 activity and a reduction in CXCR2 and VLA-4 expression on neutrophils and a decrease in VCAM-1 expression on endothelial cells.


Assuntos
Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Indóis/farmacologia , Neutrófilos/metabolismo , Animais , Bleomicina/farmacologia , Quimiotaxia/efeitos dos fármacos , Quimiotaxia de Leucócito/efeitos dos fármacos , Células Endoteliais/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Indóis/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/fisiopatologia , Transdução de Sinais/efeitos dos fármacos
8.
J Biomed Sci ; 26(1): 46, 2019 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-31189465

RESUMO

BACKGROUND: Triggering receptor expressed on myeloid cells-1 (TREM-1) is highly expressed on macrophages in inflamed intestines and reportedly promotes inflammatory bowel disease (IBD) by augmenting pro-inflammatory responses. To study the mechanism mediated by TREM-1 on macrophages, we generated an independent TREM-1 deficient mouse. METHODS: Acute colitis was induced in C57BL/6 and TREM-1-deficient mice by the administration of dextran sodium sulfate (DSS). Colonic lamina propria immune cell composition and cytokines were analyzed. An innate lymphoid cell (ILC) co-culture experiment with macrophages was used to analyze IL-22 levels. Exogenous IL-22 and TREM-1-expressing macrophages were supplied to TREM-1-deficient mice for examining their effects on intestinal barrier integrity. RESULTS: In inflamed colons, TREM-1 loss compromised the activation of ILC3 and their production of IL-22, which is required for intestinal barrier integrity. ILC3-mediated IL-22 production depends on IL-1ß secreted by M1-polarized macrophages, and we found that TREM-1 deficiency results in a decreased number of IL-1ß producing-M1 macrophages in colons exposed to DSS. Accordingly, DSS-mediated damage was ameliorated by supplying exogenous IL-22 and TREM-1-expressing macrophages to TREM-1-deficient mice. CONCLUSIONS: TREM-1 plays a crucial role in regulating IL-22 production by ILC3 through modulating M1-macrophage polarization during DSS-induced acute colitis.


Assuntos
Colite/patologia , Interleucinas/metabolismo , Mucosa Intestinal/fisiologia , Linfócitos/imunologia , Macrófagos/fisiologia , Receptor Gatilho 1 Expresso em Células Mieloides/metabolismo , Animais , Colite/induzido quimicamente , Colite/fisiopatologia , Sulfato de Dextrana/toxicidade , Mucosa Intestinal/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Interleucina 22
9.
J Autoimmun ; 78: 92-100, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28089248

RESUMO

Sensing of nucleic acids by pattern recognition receptors is the key for the initiation and development of systemic lupus erythematosus (SLE). Triggering receptor expressed on myeloid cells-1 (TREM-1) is a novel innate immune receptor, which can amplify Toll-like receptor (TLR)-induced inflammatory responses. Although patients with lupus exhibit increased serum levels of soluble TREM-1 (sTREM-1), the role of TREM-1 in SLE remains unknown. In current study, we found serum sTREM-1 levels were significantly increased in lupus patients and positively correlated with disease activity. Additionally, diseased B6.lpr mice had elevated TREM-1 in the serum, spleen, and lymph nodes. To investigate the role of TREM-1 in lupus, we established Trem-1-/-.lpr mice. Trem-1-/-.lpr mice exhibited lower survival rates and more severe lupus symptoms, including elevated proteinuria, serum anti-dsDNA antibody levels, renal immune complex depositions and lymphocyte subpopulation expansions in both the spleen and lymph nodes. Besides, Trem-1-/-.lpr mice expressed higher serum B cell-activating factor (BAFF) levels and lymph node dendritic cells (DCs) were the major source of increased BAFF. Activation of membrane-bound TREM-1 could suppress TLR9-induced BAFF expression in bone marrow-derived DCs of B6.lpr mice. Moreover, levels of sTREM-1, which could act as an antagonist of membrane-bound TREM-1, were positively correlated with levels of BAFF in the sera of lupus patients. Our findings suggest a novel modulatory role of TREM-1 in the pathogenesis of SLE. sTREM-1 production is a useful diagnostic marker and a molecular target for combination therapy of lupus.


Assuntos
Fator Ativador de Células B/biossíntese , Lúpus Eritematoso Sistêmico/etiologia , Lúpus Eritematoso Sistêmico/metabolismo , Receptor Gatilho 1 Expresso em Células Mieloides/deficiência , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Criança , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Regulação da Expressão Gênica , Humanos , Lúpus Eritematoso Sistêmico/patologia , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Mutação , Especificidade de Órgãos , Índice de Gravidade de Doença , Receptor Gatilho 1 Expresso em Células Mieloides/sangue , Receptor Gatilho 1 Expresso em Células Mieloides/genética , Receptor Gatilho 1 Expresso em Células Mieloides/metabolismo , Adulto Jovem
10.
Immunity ; 29(4): 615-27, 2008 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-18835195

RESUMO

Fas is highly expressed in activated and germinal center (GC) B cells but can potentially be inactivated by misguided somatic hypermutation. We employed conditional Fas-deficient mice to investigate the physiological functions of Fas in various B cell subsets. B cell-specific Fas-deficient mice developed fatal lymphoproliferation due to activation of B cells and T cells. Ablation of Fas specifically in GC B cells reproduced the phenotype, indicating that the lymphoproliferation initiates in the GC environment. B cell-specific Fas-deficient mice also showed an accumulation of IgG1(+) memory B cells expressing high amounts of CD80 and the expansion of CD28-expressing CD4(+) Th cells. Blocking T cell-B cell interaction and GC formation completely prevented the fatal lymphoproliferation. Thus, Fas-mediated selection of GC B cells and the resulting memory B cell compartment is essential for maintaining the homeostasis of both T and B lymphocytes.


Assuntos
Linfócitos B/imunologia , Centro Germinativo/imunologia , Linfócitos T/imunologia , Receptor fas/metabolismo , Animais , Antígenos CD/imunologia , Antígenos CD/metabolismo , Linfócitos B/metabolismo , Antígeno B7-1/imunologia , Antígeno B7-1/metabolismo , Antígenos CD28/imunologia , Antígenos CD28/metabolismo , Antígenos CD40/imunologia , Antígenos CD40/metabolismo , Antígeno CTLA-4 , Comunicação Celular , Diferenciação Celular , Proliferação de Células , Citocinas/sangue , Centro Germinativo/metabolismo , Homeostase , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Linfócitos T/metabolismo , Receptor fas/deficiência , Receptor fas/imunologia
11.
Blood ; 121(1): 95-106, 2013 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-23152543

RESUMO

Persistent high fever is one of the most typical clinical symptoms in dengue virus (DV)-infected patients. However, the source of endogenous pyrogen (eg, IL-1ß) and the signaling cascade leading to the activation of inflammasome and caspase-1, which are essential for IL-1ß and IL-18 secretion, during dengue infection have not been elucidated yet. Macrophages can be polarized into distinct phenotypes under the influence of GM-CSF or M-CSF, denoted as GM-Mϕ and M-Mϕ, respectively. We found that DV induced high levels of IL-1ß and IL-18 from GM-Mϕ (inflammatory macrophage) and caused cell death (pyroptosis), whereas M-Mϕ (resting macrophage) did not produce IL-1ß and IL-18 on DV infection even with lipopolysaccharide priming. This observation demonstrates the distinct responses of GM-Mϕ and M-Mϕ to DV infection. Moreover, up-regulation of pro-IL-1ß, pro-IL-18, and NLRP3 associated with caspase-1 activation was observed in DV-infected GM-Mϕ, whereas blockade of CLEC5A/MDL-1, a C-type lectin critical for dengue hemorrhagic fever and Japanese encephalitis virus infection, inhibits NLRP3 inflammasome activation and pyrotopsis in GM-Mϕ. Thus, DV can activate NLRP3 inflammasome via CLEC5A, and GM-Mϕ plays a more important role than M-Mϕ in the pathogenesis of DV infection.


Assuntos
Vírus da Dengue/fisiologia , Dengue/imunologia , Inflamassomos/imunologia , Lectinas Tipo C/fisiologia , Ativação de Macrófagos , Macrófagos/imunologia , Receptores de Superfície Celular/fisiologia , Apoptose , Permeabilidade Capilar , Proteínas de Transporte/imunologia , Inibidores de Caspase/farmacologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Dengue/genética , Endotélio Vascular/fisiologia , Febre/etiologia , Febre/fisiopatologia , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Interleucina-18/biossíntese , Interleucina-18/genética , Interleucina-1beta/biossíntese , Interleucina-1beta/genética , Lectinas Tipo C/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/classificação , Macrófagos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR , RNA Interferente Pequeno/farmacologia , Receptores de Superfície Celular/antagonistas & inibidores , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética , Replicação Viral
12.
Chin J Physiol ; 58(6): 367-76, 2015 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-26717915

RESUMO

TI-HU-YIN (JCKD), a compound composed of many Chinese herbs, is hypothesized to attenuate renal tubular injury and interstitial fibrosis. Moreover, its renoprotective effects were assessed in animal and in vitro studies. First, male C57BL/6 mice were under sham operation or unilateral ureteral obstruction (UUO) surgery, and then treated with phosphate buffer solution (PBS), aliskirin and valsartan (A+V), and JCKD for 14 days. At 7 and 14 days, mice were sacrificed and the kidney tissues were assessed for histopathological changes and transforming growth factor (TGF)-ß1 expression. As compared to sham group, UUO-PBS group had more serious tubular dilatation and injury, α-smooth muscle actin-positive areas, F4/80-positive macrophages, and interstitial fibrosis. Impressively, these pathologic changes were significantly attenuated in UUO mice both treated with JCKD and A+V as compared to UUO-PBS group. At 14 days, TGF-ß1 expression was significantly suppressed in kidney tissues of UUO-JCKD group as well as in UUO-A+V group. Second, TGF-ß1 production was increased in macrophage J774 cells and NRK-52E proximal tubular cells stimulated by angiotensin (Ang)-II at 10 nM for 24 h and at 1 nM for 48 h, respectively. JCKD (≥ 400 µg/ml) inhibited the TGF-ß1 production at baseline and stimulated by Ang II in both cell lines. Our study showed that JCKD reduced renal injury, macrophage infiltration and interstitial fibrosis possibly through suppressing the TGF-ß1 expression in UUO mice. Accordingly, JCKD is potential to retard the progression of chronic kidney disease. Further studies are needed to validate its renoprotective effects in the inhibition of TGF-ß1 expression and the amelioration of renal fibrosis.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Túbulos Renais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Obstrução Ureteral/tratamento farmacológico , Angiotensina II/farmacologia , Animais , Células Cultivadas , Medicamentos de Ervas Chinesas/uso terapêutico , Fibrose , Túbulos Renais/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistema Renina-Angiotensina/fisiologia , Fator de Crescimento Transformador beta1/análise
13.
J Cell Mol Med ; 18(7): 1344-57, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24758719

RESUMO

Toll-like receptors (TLR) recognize pathogens and trigger the production of vigorous pro-inflammatory cytokines [such as tumour necrosis factor (TNF)] that induce systemic damages associated with sepsis and chronic inflammation. Cooperation between signals of TLR and TNF receptor has been demonstrated through the participation of TNF receptor 1 (TNFR) adaptors in endotoxin tolerance. Here, we identify a TLR2-mediated synergy, through a MyD88-independent crosstalk, which enhances subsequent TNF-mediated nuclear factor-kappa B activation and interleukin-6 induction. Membrane-associated adaptor MAL conduces the link between TNF receptor-associated factor 6 (TRAF6) and TNFR-associated death domain, leading to a distinctive K63-ubiquitinylated TRAF6 recruitment into TNFR complex. In summary, our results reveal a novel route of TLR signal that synergistically amplifies TNF-mediated responses, indicating an innovative target for inflammation manipulation.


Assuntos
Regulação da Expressão Gênica , Interleucina-6/metabolismo , Fator 88 de Diferenciação Mieloide/fisiologia , Proteína de Domínio de Morte Associada a Receptor de TNF/fisiologia , Receptor 2 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Animais , Western Blotting , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Humanos , Imunoprecipitação , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Interleucina-6/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 2 Toll-Like/genética
14.
Infect Immun ; 82(3): 1335-42, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24396044

RESUMO

Klebsiella pneumoniae liver abscess (KPLA) is prevalent in East Asia. Liver abscess can develop after translocation of K. pneumoniae from a patient's bowel into the liver via the portal circulation. TREM-1 (triggering receptor expressed on myeloid cells 1) amplifies inflammatory signaling during infection, but its role in KPLA is poorly understood. We used an animal study to characterize the role of TREM-1 in KPLA. We compared survival rates, bacterial burdens in tissues, inflammatory cytokine levels, and histology findings between wild-type and Trem-1 knockout (KO) mice after oral inoculation of capsular type K1 K. pneumoniae. Translocation of K. pneumoniae to mesenteric lymph nodes and liver was examined, and intestinal permeability, antimicrobial peptide expression, and the clearance of K. pneumoniae in the small intestine were determined. In the absence of TREM-1, KPLA model mice showed increased K. pneumoniae dissemination, enhanced liver and systemic inflammation, and reduced survival. Impaired bacterial clearance in the small intestine causes enhanced K. pneumoniae translocation, which renders Trem-1 KO mice more susceptible to K. pneumoniae oral infection. In conclusion, TREM-1-mediated bacterial clearance in the small intestine is an important immune response against K. pneumoniae. TREM-1 deficiency enhances K. pneumoniae translocation in the small intestine and increases mortality rates in mice with KPLA.


Assuntos
Infecções por Klebsiella/imunologia , Klebsiella pneumoniae/imunologia , Abscesso Hepático/imunologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Animais , Translocação Bacteriana/genética , Translocação Bacteriana/imunologia , Citocinas/genética , Citocinas/imunologia , Modelos Animais de Doenças , Inflamação/imunologia , Inflamação/microbiologia , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Infecções por Klebsiella/genética , Infecções por Klebsiella/microbiologia , Fígado/imunologia , Fígado/microbiologia , Abscesso Hepático/genética , Abscesso Hepático/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/microbiologia , Receptor Gatilho 1 Expresso em Células Mieloides , Regulação para Cima/genética , Regulação para Cima/imunologia
15.
Kidney Int ; 86(6): 1174-86, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24918157

RESUMO

Chronic kidney disease (CKD) is an emerging worldwide public health problem. Inflammatory cell infiltration and activation during the early stages in injured kidneys is a common pathologic feature of CKD. Here, we determined whether an important inflammatory regulator, triggering receptor expressed on myeloid cells (TREM)-1, is upregulated in renal tissues collected from mouse ureteral obstruction-induced nephritis. TREM-1 is crucial for modulating macrophage polarization, and has a pivotal role in mediating tubular injury and interstitial collagen deposition in obstructive nephritis. Lysates from nephritic kidneys triggered a TREM-1-dependent M1 polarization ex vivo, consistent with the observation that granulocyte-macrophage colony-stimulating factor (GM-CSF)-derived M1 macrophages express higher levels of TREM-1 in comparison with M-CSF-derived cells. Moreover, agonistic TREM-1 cross-link significantly strengthens the inductions of iNOS and GM-CSF in M1 cells. These observations are validated by a strong clinical correlation between infiltrating TREM-1-expressing/iNOS-positive macrophages and renal injury in human obstructive nephropathy. Thus, TREM-1 may be a potential diagnostic and therapeutic target in human kidney disease.


Assuntos
Polaridade Celular , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Macrófagos/fisiologia , Glicoproteínas de Membrana/metabolismo , Nefrite/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Receptores Imunológicos/metabolismo , Obstrução Ureteral/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Nefrite/etiologia , Nefrite/patologia , RNA Mensageiro/metabolismo , Receptores Imunológicos/genética , Receptor Gatilho 1 Expresso em Células Mieloides , Regulação para Cima , Obstrução Ureteral/complicações , Obstrução Ureteral/patologia
16.
J Immunol ; 188(5): 2464-71, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22287720

RESUMO

Tumor-associated macrophages (TAMs) are the major component of tumor-infiltrating leukocytes. TAMs are heterogeneous, with distinct phenotypes influenced by the microenvironment surrounding tumor tissues. Decoy receptor 3 (DcR3), a member of the TNFR superfamily, is overexpressed in tumor cells and is capable of modulating host immunity as either a neutralizing decoy receptor or an effector molecule. Upregulation of DcR3 has been observed to correlate with a poor prognosis in various cancers. However, the mechanisms underlying the DcR3-mediated tumor-promoting effect remain unclear. We previously demonstrated that DcR3 modulates macrophage activation toward an M2-like phenotype in vitro and that DcR3 downregulates MHC class II expression in TAMs via epigenetic control. To investigate whether DcR3 promotes tumor growth, CT26-DcR3 stable transfectants were established. Compared with the vector control clone, DcR3-transfectants grew faster and resulted in TAM infiltration. We further generated CD68 promoter-driven DcR3 transgenic (Tg) mice to investigate tumor growth in vivo. Compared with wild-type mice, macrophages isolated from DcR3-Tg mice displayed higher levels of IL-10, IL-1ra, Ym1, and arginase activity, whereas the expression of IL-12, TNF-α, IL-6, NO, and MHC class II was downregulated. Significantly enhanced tumor growth and spreading were observed in DcR3-Tg mice, and the enhanced tumor growth was abolished by arginase inhibitor N-ω-hydroxy-l-norarginine and histone deacetylase inhibitor sodium valproate. These results indicated that induction of TAMs is an important mechanism for DcR3-mediated tumor progression. Our findings also suggest that targeting DcR3 might help in the development of novel treatment strategies for tumors with high DcR3 expression.


Assuntos
Diferenciação Celular/imunologia , Progressão da Doença , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/patologia , Membro 6b de Receptores do Fator de Necrose Tumoral/fisiologia , Regulação para Cima/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Feminino , Humanos , Ativação de Macrófagos/imunologia , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Camundongos Transgênicos
17.
Proc Natl Acad Sci U S A ; 108(45): 18354-9, 2011 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-22042853

RESUMO

TNF receptor-associated factor 2 (TRAF2) is a key intracellular signaling mediator that acts downstream of not only TNFα but also various members of the TNFα superfamily. Here, we report that, despite their lack of TNFα signaling, TRAF2(-/-)TNFα(-/-) mice develop an inflammatory disorder characterized by autoantibody accumulation and organ infiltration by T cells with the phenotypes of activated, effector, and memory cells. RAG1(-/-) mice reconstituted with TRAF2(-/-)TNFα(-/-) bone marrow cells showed increased numbers of hyperactive T cells and rapidly developed progressive and eventually lethal inflammation. No inflammation was observed in RAG1(-/-) mice reconstituted with TRAF2(-/-)TNFα(-/-)T-cell receptor ß(-/-) or TRAF2(-/-)TNFα(-/-)NFκB-induced kinase(+/-) bone marrow cells. The pathogenic TRAF2(-/-)TNFα(-/-) T cells showed constitutive NFκB2p52 activation and produced elevated levels of T-helper 1 and T-helper 17 cytokines. Our results suggest that a regulatory circuit consisting of TRAF2-NFκB-induced kinase-NFκB2p52 is essential for the proper control of effector T-cell polarization and that loss of T-cell TRAF2 function induces constitutive NFκB2p52 activity that drives fatal autoimmune inflammation independently of TNFα signaling. The involvement of this regulatory circuit in controlling autoimmune responses highlights the delicate balance required to avoid paradoxical adverse events when implementing new targeted anti-inflammatory therapies.


Assuntos
Autoimunidade , NF-kappa B/metabolismo , Transdução de Sinais , Fator 2 Associado a Receptor de TNF/fisiologia , Animais , Western Blotting , Citocinas/biossíntese , Citometria de Fluxo , Inflamação/fisiopatologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase
18.
Nephrol Dial Transplant ; 28(10): 2477-83, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24078640

RESUMO

BACKGROUND: Melamine and cyanuric acid (M/CA), when orally administered together to rats, can induce crystal formation within renal tubules and cause acute kidney injury. METHODS: To investigate the pathomechanism of crystal-induced nephritis, melamine and/or cyanuric acid were administered to 3-week-old (young) and 8-week-old (adult) rats, respectively. RESULTS: Crystal formation, blood urea nitrogen elevation, tubular cell injury and macrophage infiltration were noted in rats fed with M/CA, but not in rats fed with vehicle, melamine or CA alone. These parameters were significantly higher in young rats than those in adult rats fed with M/CA 200 mg/kg body weight (BW) for 3 days. Krüppel-like factor 5 (KLF5) was expressed on distal tubule cells, especially when crystals deposited within the lumens. Both mRNA and protein levels were higher in young rats than those in adult rats fed with M/CA (200 mg/kg BW). KLF5 expression has been shown to modulate renal tissue cytokine production, and we found that proinflammatory cytokines like monocyte chemoattractant protein-1 and interlukin-6 were increased in kidney tissues of young rats fed with M/CA for 3 days. In contrast, interlukin-10, an anti-inflammatory cytokine, was upregulated in kidneys of adult rats fed with M/CA for 3 days. CONCLUSIONS: Crystals are prone to deposition in distal tubules of young rats fed with M/CA. M/CA Crystal-related nephritis might be induced by the KLF5 expression, which modulated macrophage recruitment and proinflammatory cytokine production, subsequently leading to renal tubular injury and interstitial inflammation.


Assuntos
Inflamação/patologia , Túbulos Renais/lesões , Fatores de Transcrição Kruppel-Like/metabolismo , Nefrite/patologia , Triazinas/toxicidade , Animais , Western Blotting , Técnicas Imunoenzimáticas , Inflamação/etiologia , Inflamação/metabolismo , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Masculino , Nefrite/induzido quimicamente , Nefrite/metabolismo , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Resinas Sintéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Nature ; 446(7132): 203-7, 2007 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-17322907

RESUMO

Complement-derived anaphylatoxins regulate immune and inflammatory responses through G-protein-coupled receptor (GPCR)-mediated signalling. C5L2 (also known as GPR77) is a relatively new GPCR thought to be a non-signalling receptor binding to C5a, on the basis of sequence information and experimental evidence. Here we show, using gene targeting, that C5L2 is required to facilitate C5a signalling in neutrophils, macrophages and fibroblasts in vitro. Deficiency of C5L2 results in reduced inflammatory cell infiltration, suggesting that C5L2 is critical for optimal C5a-mediated cell infiltration in certain in vivo settings. C5L2 is also involved in optimizing C3a-induced signals. Furthermore, like mice incapable of C3a/complement 3a receptor (C3aR) signalling, C5L2-deficient mice are hypersensitive to lipopolysaccharide (LPS)-induced septic shock, show reduced ovalbumin (OVA)-induced airway hyper-responsiveness and inflammation, and are mildly delayed in haematopoietic cell regeneration after gamma-irradiation. Our data indicate that C5L2 can function as a positive modulator for both C5a- and C3a-anaphylatoxin-induced responses.


Assuntos
Complemento C3a/metabolismo , Complemento C5a/metabolismo , Actinas/química , Actinas/metabolismo , Animais , Bovinos , Células Cultivadas , Ativação do Complemento , Complemento C3a/imunologia , Complemento C5a/imunologia , Fibroblastos , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Humanos , Inflamação , Pulmão/citologia , Neutrófilos/citologia , Neutrófilos/metabolismo , Receptor da Anafilatoxina C5a , Receptores de Quimiocinas/deficiência , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Transdução de Sinais
20.
Exp Hematol Oncol ; 12(1): 35, 2023 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-37029450

RESUMO

INTRODUCTION: Circulating monocytic myeloid-derived suppressive cells (M-MDSCs) are implicated as a poor prognostic factor and cause CAR T-cell failure in diffuse large B-cell lymphoma (DLBCL). Triggering receptors expressed on myeloid cells 2 (TREM2) are a transmembrane glycoprotein that polarize macrophages to anti-inflammation phenotype but have never been explored on M-MDSCs. This study aims to elucidate the expression and clinical impact of surface TREM2 on circulating M-MDSCs derived from DLBCL adults. METHODS: This prospective, observational study enrolled 100 adults with newly diagnosed and treatment-naïve DLBCL from May 2019 to October 2021. Human circulating M-MDSCs were obtained from freshly isolated peripheral blood, and each patient's surface-TREM2 level on M-MDSCs was normalized via a healthy control at the same performance of flow-cytometry analysis. Murine MDSCs derived from bone marrow (BM-MDSCs) were adopted to assess the link between Trem2 and cytotoxic T lymphocytes. RESULTS: More circulating M-MDSCs at diagnosis of DLBCL predicted worse progression-free (PFS) and overall survival (OS). Patients with higher IPI scores, bone marrow involvement, or lower absolute counts of CD4+ or CD8+ T cells in PB had significantly higher normalized TREM2 levels on M-MDSCs. Additionally, normalized TREM2 levels on M-MDSCs could be grouped into low (< 2%), medium (2-44%), or high (> 44%) levels, and a high normalized TREM2 level on M-MDSCs was proven as an independent prognostic factor for both PFS and OS via multivariate Cox regression analysis and associated with worst PFS and OS. Interestingly, normalized levels of surface TREM2 on M-MDSCs were negatively associated with absolute counts of PB CD8+ T cells and positively correlated with levels of intracellular arginase 1 (ARG1) within M-MDSCs. Wild-type BM-MDSCs had significantly higher mRNA levels of Arg1 and showed more prominent ability to suppress the proliferation of co-cultured CD8+ T cells than BM-MDSCs from Trem2 knockout mice, and the suppressive ability could be impaired by adding Arg1 inhibitors (CB1158) or supplementing L-arginine. CONCLUSION: In treatment-naïve DLBCL adults, a high surface-TREM2 level on circulating M-MDSCs is a poor prognostic factor for both PFS and OS and warrants further investigation for its potential as a novel target in immunotherapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA