Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
World J Surg Oncol ; 21(1): 371, 2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-38008720

RESUMEN

BACKGROUND: Long non-coding RNAs (lncRNAs) are critical regulators in the initiation and progression of breast cancer. Our study aims to characterize the functions of LINC02086 which few published in breast cancer and decipher the downstream molecular mechanisms. METHODS: LINC02086 expression is tested in RNA-seq data from GEPIA database, tumor tissue samples from hospital patients and breast cancer cell lines. LINC02086 was silenced or overexpressed by lenti-virus-mediated shRNAs, or pLVX-Puro plasmids. Luciferase reporter assay and RNA pull-down assay were applied to study interactions between LINC02086, miR-6757-5p and ephrin type-A receptor 2 (EPHA2). LINC02086-silencing MCF-7 cells were injected into mice to establish xenograft animal models. RESULTS: Using RNA-seq data, tumor tissue samples and breast cancer cells, LINC02086 was consistently found to be up-regulated in breast cancer, and correlated with poorer prognosis. LINC02086 knockdown decreased cell viability, promoted cell apoptosis and suppressed tumor growth. LINC02086 interacted with miR-6757-5p that interacted with EPHA2.LINC02086 expression was negatively correlated with miR-6757-5p expression (r = -0.5698, P < 0.001) but was positively correlated with EPHA2 expression (r = 0.5061, P < 0.001). miR-6757-5p expression was negatively correlated with EPHA2 expression (r = -0.5919, P < 0.001). LINC02086 regulated EPHA2 via miR-6757-5p. miR-6757-5p/EPHA2 axis was a mediator of the effect of LINC02086 on cell viability and apoptosis. CONCLUSION: LINC02086 increases cell viability and decreases apoptotic cells in breast cancer by sponging miR-6757-5p to upregulate EPHA2. This study presents LINC02086/miR-6757-5p/EPHA2 axis as promising therapeutic targets for breast cancer intervention.


Asunto(s)
Neoplasias de la Mama , MicroARNs , ARN Largo no Codificante , Animales , Femenino , Humanos , Ratones , Apoptosis/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
2.
Biochem Biophys Res Commun ; 529(3): 839-845, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32616311

RESUMEN

Sepsis-associated encephalopathy (SAE) is a devastating neurological complication of sepsis with intolerable high motility. SAE is accompanied with brain vascular injury, endothelial hyperpermeability, and neutrophil infiltration into the brain tissue, key inflammatory processes leading to further brain edema and neuronal cell apoptosis. Recent studies from us and others suggest that the chemokine receptor C-X-C Motif Chemokine Receptor 2 (CXCR2) is crucial for neutrophil recruitment during SAE. Here we use CXCR2 antagonist SB225002 to characterize the role of CXCR2 in brain infiltration of neutrophil in a murine model of SAE. Systemic administration of high-dose LPS (10 mg/kg) induced evident neutrophil infiltration into the cerebral cortex in wild-type mice. However, CXCR2 antagonist SB225002 markedly attenuated neutrophil infiltration into brain. The CXCR2 expression on neutrophils in the peripheral circulation was dramatically downregulated in response to this LPS dose, and endothelial CXCR2 was significantly upregulated, suggesting endothelial but not neutrophil CXCR2 plays a more important role in neutrophil infiltration into brain. Strikingly, although these CXCR2 antagonist SB225002 treated mice displayed reduced neutrophil infiltration, no change in neutrophil rolling and adhesion was observed. Furthermore, we confirmed that CXCR2 agonist CXCL1 induced a marked increase in actin stress fiber synthesis and paracellular gap formation in cultured cerebral endothelial cells, which is attenuated by SB225002. Thus, these results demonstrate a selective role for endothelial CXCR2 to regulate cerebral vascular permeability and neutrophil transmigration in high-dose LPS induced neuroinflammation, and also suggest a therapeutic potential of CXCR2 antagonist SB225002 in SAE.


Asunto(s)
Antiinflamatorios/uso terapéutico , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Compuestos de Fenilurea/uso terapéutico , Receptores de Interleucina-8B/antagonistas & inhibidores , Encefalopatía Asociada a la Sepsis/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Edema Encefálico , Línea Celular , Modelos Animales de Enfermedad , Lipopolisacáridos/inmunología , Masculino , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Receptores de Interleucina-8B/inmunología , Encefalopatía Asociada a la Sepsis/inmunología
3.
Clin Immunol ; 198: 89-99, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30502542

RESUMEN

BACKGROUND: Programmed cell death-1 (PD-1) and programmed death-ligand 1 (PD-L1)-targeted therapies have enhanced T-cell response and demonstrated efficacy in the treatment of multiple cancers. However, the role and clinical significance of PD-L1 expression on CD19+ B-cells and their subsets, with particular reference to systemic lupus erythematosus (SLE), have not yet been studied in detail. OBJECTIVE: The present study aimed to investigate PD-L1 expression on CD19+ B-cells and their subsets, in addition to exploring its possible role in Tfh-cell activation and B-cell differentiation in SLE. METHODS: Frequencies of CD19+ B-cells, their subsets, PD-L1 and Tfh cells in the peripheral blood of SLE patients and healthy controls (HCs) were determined using cytometry. The clinical data of SLE patients were recorded in detail, and the correlation between their laboratory parameters, clinical parameters and disease activity indices was statistically analyzed. CD19+PD-L1+B-cells and CD19+PD-L1- B-cells were sorted and cultured with a stimulant, following which the supernatants were collected for immunoglobulin G and anti-double stranded DNA detection via enzyme-linked immunosorbent assay. RESULTS: In SLE patients, CD19+B-cells and partial subgroups were enriched in peripheral blood. Also, the observed increase in the frequency of CD19+PD-L1+B-cells was significantly associated with a higher disease activity index. An in vitro culture test demonstrated that the amounts of anti-dsDNA and immunoglobulin G secreted by the CD19+PD-L1+B-cells of SLE patients and HCs were vastly different. In addition, a strong correlation existed between the frequencies of CD19+PD-L1+B-cells and defined Tfh cells of SLE patients. CONCLUSION: This study demonstrated that the expression of CD19+PD-L1+B-cells in the peripheral blood of SLE patients was abnormal, and that disease-related laboratory parameters and clinical indicators were correlated. CD19+PD-L1+B-cells were enriched and played a critical role in activating the pathogenic T-cell and B-cell responses in patients with SLE.


Asunto(s)
Antígenos CD19/análisis , Linfocitos B/inmunología , Antígeno B7-H1/fisiología , Lupus Eritematoso Sistémico/inmunología , Adulto , ADN/inmunología , Femenino , Humanos , Inmunoglobulina G/biosíntesis , Masculino , Persona de Mediana Edad , Linfocitos T Colaboradores-Inductores/inmunología
4.
Cent Eur J Immunol ; 42(1): 30-38, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28680329

RESUMEN

miR-20b is a member of the miR-106a-363 gene cluster, which has been shown to play an important role in a variety of diseases, including cancer, inflammation, and autoimmune diseases. Our previous study indicated that miR-20b has an inhibitory effect on airway inflammation in asthmatic mice, but the exact mechanism is unclear. In this study, we report that the ratio of CD11b+Ly6G+Ly6Clow cells, but not the amount of CD11b+Ly6C+Ly6G- cells, was increased in the lung tissue of asthmatic mice after intranasal instillation with miR-20b mimics, while Th2-type cytokines (interleukin (IL)-4 and IL-13) were significantly decreased in the bronchoalveolar lavage fluid. In addition, the transcription factor CREB regulated the expression of miR-20b. Our findings suggest that miR-20b can induce the accumulation of myeloid-derived suppressor cells in the lungs of asthmatic mice, which may be a mechanism by which miR-20b inhibits airway inflammation in asthmatic mice. Thus, miR-20b may be used as a target for the effective treatment of asthma in the future.

5.
Oncol Lett ; 27(6): 256, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38646501

RESUMEN

Cluster of differentiation 47 (CD47) is a transmembrane protein that is widely and moderately expressed on the surface of various cells and can have an essential role in mediating cell proliferation, migration, phagocytosis, apoptosis, immune homeostasis and other related responses by binding to its ligands, integrins, thrombospondin-1 and signal regulatory protein α. The poor prognosis of cancer patients is closely associated with high expression of CD47 in glioblastoma, ovarian cancer, breast cancer, bladder cancer, colon cancer and hepatocellular carcinoma. Upregulation of CD47 expression facilitates the growth of numerous types of tumor cells, while downregulation of its expression promotes phagocytosis of tumor cells by macrophages, thereby limiting tumor growth. In addition, blocking CD47 activates the cyclic GMP-AMP (cGAMP) synthase/cGAMP/interferon gene stimulating factor signaling pathway and initiates an adaptive immune response that kills tumor cells. The present review describes the structure, function and interactions of CD47 with its ligands, as well as its regulation of phagocytosis and tumor cell fate. It summarizes the therapeutics, mechanisms of action, research advances and challenges of targeting CD47. In addition, this paper provides an overview of the latest therapeutic options for targeting CD47, such as chimeric antigen receptor (CAR) T-cells, CAR macrophages and nanotechnology-based delivery systems, which are essential for future clinical research on targeting CD47.

6.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(1): 13-18, 2024 Jan.
Artículo en Zh | MEDLINE | ID: mdl-38246172

RESUMEN

Objective To investigate the effect of interleukin-6 (IL-6) on the phagocytosis of MH-S alveolar macrophages and its related mechanisms. Methods A mouse acute lung injury (ALI) model was constructed by instilling lipopolysaccharide (LPS) into the airway. ELISA was used to detect the content of IL-6 in bronchoalveolar lavage fluid (BALF). In vitro cultured MH-S cells, in the presence or absence of signal transducer and activator 3 of transcription(STAT3) inhibitor Stattic (5 µmol/L), IL-6 (10 ng/mL~500 ng/mL) was added to stimulate for 6 hours, and then incubated with fluorescent microspheres for 2 hours. The phagocytosis of MH-S cells was detected by flow cytometry. Western blot analysis was used to detect the expression levels of phosphorylated Janus kinase 2 (p-JAK2), phosphorylated STAT3 (p-STAT3), actin-related protein 2 (Arp2) and filamentous actin (F-actin). Results The content of IL-6 in BALF was significantly increased after the mice were injected with LPS through the airway. With the increase of IL-6 stimulation concentration, the phagocytic function of MH-S cells was enhanced, and the expression levels of Arp2 and F-actin proteins in MH-S cells were increased. The expression levels of p-JAK2 and p-STAT3 proteins increased in MH-S cells stimulated with IL-6(100 ng/mL). After blocking STAT3 signaling, the effect of IL-6 in promoting phagocytosis of MH-S cells disappeared completely, and the increased expression of Arp2 and F-actin proteins in MH-S cells induced by IL-6 was also inhibited. Conclusion IL-6 promotes the expression of Arp2 and F-actin proteins by activating the JAK2/STAT3 signaling pathway, thereby enhancing the phagocytic function of MH-S cells.


Asunto(s)
Interleucina-6 , Macrófagos Alveolares , Animales , Ratones , Actinas , Modelos Animales de Enfermedad , Janus Quinasa 2 , Lipopolisacáridos , Transducción de Señal
7.
Mol Immunol ; 166: 58-64, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38244369

RESUMEN

Surfactant protein A (SP-A), a natural immune molecule, plays an important role in lung health. SP-A recognizes and binds microbial surface glycogroups through the C-type carbohydrate recognition domain, and then binds corresponding cell surface receptors (such as C1qRp, CRT-CD91 complex, CD14, SP-R210, Toll-like receptor, SIRP-α, CR3, etc.) through collagen-like region, and subsequently mediates biological effects. SP-A regulates lung innate immunity by promoting surfactant absorption by alveolar type II epithelial cells and phagocytosis of pathogenic microorganisms by alveolar macrophages. SP-A also regulates lung adaptive immunity by inhibiting DC maturation, and T cell proliferation and differentiation. This article reviews latest relationships between SP-A and adaptive and intrinsic immunity.


Asunto(s)
Macrófagos Alveolares , Proteína A Asociada a Surfactante Pulmonar , Proteína A Asociada a Surfactante Pulmonar/metabolismo , Pulmón/metabolismo , Fagocitosis , Inmunidad Innata , Proteína D Asociada a Surfactante Pulmonar
8.
Artículo en Inglés | MEDLINE | ID: mdl-38726776

RESUMEN

BACKGROUND: Breast Cancer (BC) is a female malignancy with a high mortality rate. Novel diagnostic and prognostic biomarkers are valuable for reducing BC mortality. Our study is designed to undrape the precise role of the LINC00466/miR-4731-5p/EPHA2 axis in BC.

Methods: The Cancer Genome Atlas (TCGA) sequencing dataset was utilized to compare the levels of LINC00466. The levels of LINC00466, miR-4731-5p, and EPHA2 were tested by qRTPCR. Cell proliferation and cycle were detected by CCK-8 assay and flow cytometer. In vivo role of LINC00466 was tested by Xenograft nude models. Binding sites were predicted by TargetScan and Starbase. The binding relationship was employed by Dual-luciferase reporter gene assay and RNA pull-down assay.

Results: LINC00466 was increased in human breast cancer tissues. LINC00466 was negatively associated with miR-4731-5p and positively correlated with EPHA2 in human breast cancer tissues. Down-regulation of LINC00466 suppressed the proliferation and arrested the cell cycle of breast cancer cells, and inhibited tumor growth in vivo.

Conclusion: LINC00466 promoted BC development via mediating the miR-4731-5p/EPHA2 axis, which has the potential value as a promising therapeutic target in BC.

9.
Front Immunol ; 14: 1158859, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37081870

RESUMEN

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic growth factor originally identified as a stimulus that induces the differentiation of bone marrow progenitor cells into granulocytes and macrophages. GM-CSF is now considered to be a multi-origin and pleiotropic cytokine. GM-CSF receptor signals activate JAK2 and induce nuclear signals through the JAK-STAT, MAPK, PI3K, and other pathways. In addition to promoting the metabolism of pulmonary surfactant and the maturation and differentiation of alveolar macrophages, GM-CSF plays a key role in interstitial lung disease, allergic lung disease, alcoholic lung disease, and pulmonary bacterial, fungal, and viral infections. This article reviews the latest knowledge on the relationship between GM-CSF and lung balance and lung disease, and indicates that there is much more to GM-CSF than its name suggests.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos , Pulmón , Humanos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Pulmón/metabolismo , Enfermedades Pulmonares Intersticiales , Macrófagos Alveolares , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
10.
J Ovarian Res ; 16(1): 171, 2023 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-37612724

RESUMEN

The functions and molecular mechanisms of long noncoding RNA (lncRNA) in reproduction have been widely studied at present. However, lncRNA regulating hormone synthesis in ovarian follicular granulosa cells has not been sufficiently studied. Our previous research demonstrated that lncRNA Gm2044 could promote estradiol synthesis in follicular granulosa cells. In this study, we identified 21 binding proteins of lncRNA Gm2044 in ovarian follicles using comprehensive identification of RNA-binding proteins by mass spectrometry (ChIRP-MS). RNA immunoprecipitation (RNA IP) and reverse transcription PCR (RT-PCR) confirmed that lncRNA Gm2044 can interact with eukaryotic translation elongation factor 2 (EEF2) protein. Furthermore, we constructed lncRNA Gm2044 knockout mice using the CRISPR/Cas9 method. Although the follicular development and fertility of female lncRNA Gm2044 knockout mice were not affected, the serum estradiol concentration in female lncRNA Gm2044 knockout mice significantly decreased. Western blotting and ELISA revealed that lncRNA Gm2044 may promote the binding of EEF2 to Nr5a1 mRNA and then enhance the Nr5a1 mRNA translation, and the upregulated NR5A1 protein can strengthen estradiol synthesis. To determine the potential signaling pathway of lncRNA Gm2044 regulating estradiol synthesis, transcriptome sequencing was performed for ovaries of adult lncRNA Gm2044 knockout mice, which identified 565 significant up-regulated genes and 303 significant down-regulated genes, which were then analyzed with Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) and validated by molecular experiments. Understanding how lncRNA Gm2044/EEF2 protein regulates estradiol synthesis will help treat estrogen-related reproductive diseases.


Asunto(s)
Estradiol , ARN Largo no Codificante , Femenino , Animales , Ratones , ARN Largo no Codificante/genética , Factor 2 de Elongación Peptídica , Células de la Granulosa , Ratones Noqueados
11.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 39(2): 117-123, 2023 Feb.
Artículo en Zh | MEDLINE | ID: mdl-36872429

RESUMEN

Objective To investigate the effects of C-X-C motif chemokine ligand 1 (CXCL1) and its receptor CXCR2 on the cerebral endothelial cytoskeleton rearrangement and permeability in the inflammation of septic encephalopathy. Methods The murine model of septic encephalopathy was established by intraperitoneal injection of LPS (10 mg/kg). The levels of TNF-α and CXCL1 in the whole brain tissue were detected by ELISA. The expression of CXCR2 was detected by Western blot analysis after bEND.3 cells were stimulated with 500 ng/mL LPS and 200 ng/mL TNF-α. After treated with CXCL1(150 ng/mL), the changes of endothelial filamentous actin (F-actin) rearrangement in bEND.3 cells were observed by immuno-fluorescence staining. In the cerebral endothelial permeability test, bEND.3 cells were randomly divided into PBS control group, CXCL1 group, and CXCL1 combined with CXCR2 antagonist SB225002 group. Then endothelial transwell permeability assay kit was used to detect the endothelial permeability changes. After stimulated with CXCL1 in bEND.3 cells, Western blot analysis was used to detect the expression of protein kinase B (AKT) and phosphorylated-AKT (p-AKT). Results Intraperitoneal injection of LPS significantly increased the levels of TNF-α and CXCL1 in the whole brain. LPS and TNF-α both upregulated the expression of CXCR2 protein in bEND.3 cells. CXCL1 stimulation induced the endothelial cytoskeleton contraction, increased paracellular gap formation and elevated endothelial permeability in bEND.3 cells, which was inhibited by the pretreatment with SB225002(CXCR2 antagonist). Furthermore, CXCL1 stimulation also enhanced the phosphorylation of AKT in bEND.3 cells. Conclusion CXCL1 induces the cytoskeleton contraction and increased permeability through AKT phosphorylation in bEND.3 cells, which can be effectively inhibited by CXCR2 antagonist SB225002.


Asunto(s)
Encefalopatías , Células Endoteliales , Animales , Ratones , Proteínas Proto-Oncogénicas c-akt , Fosforilación , Lipopolisacáridos , Factor de Necrosis Tumoral alfa , Citoesqueleto , Endotelio
12.
Cancer Immunol Immunother ; 61(3): 303-12, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21877248

RESUMEN

Mast cells may have either antitumor or tumor-promoting potential. Nevertheless, mast cells in tumor microenvironment have been found to promote tumor growth. So far the mechanisms underlying the modulation of mast cell function in tumor microenvironment remains to be fully elucidated. Here, we report that tumor-promoting potential of mast cells could be augmented by molecules released from damaged tumor cells through cooperative stimulation of stem cell factor (SCF) and ligand for Toll-like receptor 4 (TLR4). Co-simulation with SCF and TLR4 ligand inhibited mast cell degranulation, but efficiently induced the production and secretion of VEGF, PDGF, and IL-10. Although TLR4 ligand alone may induce IL-12 expression in mast cells, co-stimulation with SCF and TLR4 ligand induced the expression of IL-10, but not IL-12, in mast cells. The phosphorylation of GSK3ß was crucial for the effect of SCF and TLR4 ligand. In addition to inducing phosphorylation of GSK3ß at Ser9 through PI3K pathway, SCF and TLR4 ligand cooperated to induce phosphorylation of GSK3ß at Tyr216 by simultaneous activation of ERK and p38MAPK pathways. Both phospho-Ser9 and phospho-Tyr216 of GSK3ß were required for IL-10 expression induced by SCF/TLR4 ligand, whereas suppressive effect of SCF/TLR4 ligand on mast cell degranulation was related to phospho-Tyr216. Importantly, the effect of SCF and TLR4 ligand on mast cells could be abrogated by inhibiting phosphorylation of GSK3ß at Tyr216. These findings disclose the mechanisms underlying the modulation of mast cell function in tumor microenvironment, and suggest that inhibiting GSK3ß in mast cells will be beneficial to the treatment of cancer.


Asunto(s)
Mastocitos/inmunología , Neoplasias Experimentales/inmunología , Factor de Células Madre/inmunología , Receptor Toll-Like 4/inmunología , Animales , Western Blotting , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/fisiología , Degranulación de la Célula/efectos de los fármacos , Degranulación de la Célula/inmunología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Células Cultivadas , Cromolin Sódico/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-12/genética , Interleucina-12/metabolismo , Ligandos , Lipopolisacáridos/farmacología , Mastocitos/efectos de los fármacos , Mastocitos/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Fosforilación/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Células Madre/metabolismo , Factor de Células Madre/farmacología , Receptor Toll-Like 4/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
13.
J Immunol ; 185(5): 2773-82, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20675592

RESUMEN

Gr-1(+)CD11b(+)F4/80(+) cells play important roles in tumor development and have a negative effect on tumor immunotherapy. So far, the mechanisms underlying the regulation of their immunosuppressive phenotype by classical and alternative macrophage activation stimuli are not well elucidated. In this study, we found that molecules from necrotic tumor cells (NTC-Ms) stimulated Gr-1(+)CD11b(+)F4/80(+) cells to induce apoptosis of activated T cells but not nonstimulated T cells. The apoptosis-inducing capacity was determined by higher expression levels of arginase I and IL-10 relative to those of NO synthase 2 and IL-12 in Gr-1(+)CD11b(+)F4/80(+) cells, which were induced by NTC-Ms through TLR4 signaling. The apoptosis-inducing capacity of NTC-Ms-stimulated Gr-1(+)CD11b(+)F4/80(+) cells could be enhanced by IL-10. IFN-gamma may reduce the apoptosis-inducing capacity of Gr-1(+)CD11b(+)F4/80(+) cells only if their response to IFN-gamma was not attenuated. However, the potential of Gr-1(+)CD11b(+)F4/80(+) cells to express IL-12 in response to IFN-gamma could be attenuated by tumor, partially due to the existence of active STAT3 in Gr-1(+)CD11b(+)F4/80(+) cells and NTC-Ms from tumor. In this situation, IFN-gamma could not effectively reduce the apoptosis-inducing capacity of Gr-1(+)CD11b(+)F4/80(+) cells. Tumor immunotherapy with 4-1BBL/soluble programmed death-1 may significantly reduce, but not abolish the apoptosis-inducing capacity of Gr-1(+)CD11b(+)F4/80(+) cells in local microenvironment. Blockade of TLR4 signaling could further reduce the apoptosis-inducing capacity of Gr-1(+)CD11b(+)F4/80(+) cells and enhance the suppressive effect of 4-1BBL/soluble form of programmed death-1 on tumor growth. These findings indicate the relationship of distinct signaling pathways with apoptosis-inducing capacity of Gr-1(+)CD11b(+)F4/80(+) cells and emphasize the importance of blocking TLR4 signaling to prevent the induction of T cell apoptosis by Gr-1(+)CD11b(+)F4/80(+) cells.


Asunto(s)
Apoptosis/inmunología , Antígeno CD11b/biosíntesis , Carcinoma Hepatocelular/metabolismo , Factor de Crecimiento Epidérmico/biosíntesis , Neoplasias Hepáticas/metabolismo , Activación de Linfocitos/inmunología , Receptores de Quimiocina/biosíntesis , Subgrupos de Linfocitos T/inmunología , Receptor Toll-Like 4/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/fisiología , Antígeno CD11b/fisiología , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Células Cultivadas , Factor de Crecimiento Epidérmico/fisiología , Ligandos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Necrosis , Receptores de Quimiocina/fisiología , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Receptor Toll-Like 4/antagonistas & inhibidores , Receptor Toll-Like 4/fisiología
14.
Immun Inflamm Dis ; 10(7): e662, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35759236

RESUMEN

Prostaglandin E2 (PGE2) is a potent lipid mediator of inflammation that modulates immune cell function by binding to unique G protein-coupled receptors (EP receptors). PGE2 production increases during microbial infection and inflammation. In this study, we assessed the effect of PGE2 on the phagocytosis of bacteria by neutrophils, which are key players during infection and inflammation. We also looked for specific EP receptor signaling pathways that contributed to the neutrophil phagocytic activity. PGE2 (50-1000 ng/ml) inhibited the phagocytosis of Escherichia coli by HL-60 human neutrophils in a concentration-dependent manner. Inhibition of neutrophil phagocytosis by PGE2 correlated with increased intracellular cyclic adenosine monophosphate (cAMP) production, and forskolin, an adenosyl cyclase agonist, confirmed the inhibitory effect of cAMP stimulation on neutrophil phagocytosis. The expression of EP2 receptors by HL-60 cells was confirmed by western blot analysis, and selective agonism of EP2 receptors mimicked the inhibition of phagocytosis by PGE2. The EP2 receptor antagonist AH-6089 partially blocked the inhibition of neutrophil phagocytosis PGE2. Specific inhibition of phosphatase and tensin homolog (PTEN) enzyme attenuated the inhibition of neutrophil phagocytosis by PGE2, and both PGE2 and increased intracellular cAMP increased neutrophil PTEN activity, which was associated with decreased PTEN phosphorylation. The results support negative regulation of the antimicrobial activity of neutrophils (i.e., phagocytosis), which has important implications for the future management of bacterial infections.


Asunto(s)
Dinoprostona , Neutrófilos , AMP Cíclico/metabolismo , AMP Cíclico/farmacología , Dinoprostona/metabolismo , Dinoprostona/farmacología , Humanos , Inflamación , Fosfohidrolasa PTEN/farmacología , Fagocitosis , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo
15.
Free Radic Biol Med ; 193(Pt 1): 177-189, 2022 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-36244589

RESUMEN

Tuberculosis (TB), caused by mycobacterium tuberculosis (M. tuberculosis) infection, is one of the leading causes of death globally and poses a threat to public health. During infection, M. tuberculosis causes redox imbalance and dysfunctions of protective immunity. Transcription factor nuclear factor erythroid 2 (NF-E2)-related factor (Nrf2) is a major modulator of cellular redox homeostasis via transcriptional induction of cytoprotective genes to protect cell against the damage from insults. Thus, we hypothesize that Nrf2 may regulate protective immunity against M. tuberculosis. RNA-seq and immunoblotting results suggested that the expression of Nrf2 protein increased after M. tuberculosis infection, and decreased upon long-term M. tuberculosis infection, while Keap1 protein maintained a low expression level during M. tuberculosis infection. Furthermore, Nrf2 activator sulforaphane (SFN) decreased proinflammatory cytokines production, phagocytosis and host cell apoptosis, while increasing ROS levels and promoting autophagy in THP1 macrophages infected with M. tuberculosis. In addition, SFN-activated Nrf2 augmented bacterial killing by macrophages, which might be due to the regulation of protective immunity via Nrf2. Combined, our results extend the understanding of the complex innate immunity regulation by Nrf2 against mycobacterial infection. Also, these findings suggested that the regulation of Nrf2 signaling cascade could be used as a therapeutic target for the treatment of TB patients and the development of better anti-TB vaccines.


Asunto(s)
Macrófagos , Mycobacterium tuberculosis , Factor 2 Relacionado con NF-E2 , Tuberculosis , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Tuberculosis/genética , Tuberculosis/metabolismo , Células THP-1
16.
J Immunol ; 182(10): 6618-24, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19414817

RESUMEN

Pregnant women have both the local immune tolerance at the maternal-fetal interface and the systemic immune defense against pathogens. To date, regardless of the extensive investigation on the maternal-fetal immune tolerance, the maintenance of systemic immune defense in pregnant women still remains poorly understood. In the present study, we demonstrate that the immunoregulatory molecule T cell Ig and mucin domain (Tim)-3 plays important roles in innate and adaptive immunity of human pregnancy. During pregnancy, Tim-3 is strikingly up-regulated in peripheral blood of pregnant women, most by monocytes but not by T or B cells. The increased IL-4/STAT6 signaling may contribute to such up-regulation of Tim-3. In turn, the increased Tim-3 enhances not only innate immunity but also Th1-associated immune responses of pregnant women against pathogens. In contrast, our clinical data show that abnormal Tim-3 expression level might be connected to the pregnancy loss. In conclusion, our data show in this study that an immune regulatory molecule Tim-3, by virtue of its up-regulation in innate immune cells in pregnant women, enhances both innate and adaptive immune responses. Nevertheless, the abnormality of Tim-3 in pregnant woman may be deleterious to normal pregnancy.


Asunto(s)
Aborto Espontáneo/inmunología , Inmunidad Innata , Proteínas de la Membrana/metabolismo , Monocitos/inmunología , Embarazo/inmunología , Femenino , Citometría de Flujo , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Interleucina-4/inmunología , Interleucina-4/metabolismo , Proteínas de la Membrana/inmunología , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT6/inmunología , Factor de Transcripción STAT6/metabolismo , Transducción de Señal/inmunología , Células TH1/inmunología , Células Th2/inmunología , Regulación hacia Arriba
17.
Tohoku J Exp Med ; 225(1): 59-63, 2011 09.
Artículo en Inglés | MEDLINE | ID: mdl-21869592

RESUMEN

The incidence of neonatal inflammatory diseases remains high despite improved strategies for dealing with infection. Neutrophils are believed to play a significant role in neonatal inflammatory diseases due to their secretion of harmful mediators. Neutrophils rapidly undergo apoptosis following activation; dysregulation of the neutrophil apoptotic pathway may be an underlying mechanism of neonatal inflammatory disorders. In this study, we determined whether neonatal neutrophils are intrinsically resistant to apoptosis relative to their adult counterparts. Twelve healthy full-term newborn infants and 12 healthy adult volunteers, aged 20 to 45 years, were enrolled in this study. Neutrophils were isolated from umbilical cord blood or fresh venous peripheral blood, and neutrophils of each subject were cultured for up to 48 hours. Flow cytometric analysis revealed that spontaneous apoptosis of adult neutrophils increased with culture time (23% ± 2% at 12 h, 49% ± 4% at 24 h and 76% ± 3% at 48 h), whereas the frequency of apoptosis was significantly lower in neutrophils from neonates (9% ± 2% at 12 h, 30% ± 4% at 24 h and 49% ± 3% at 48 h) (p < 0.01 for each time point). Importantly, the expression levels of caspase-3 mRNA and a precursor form of caspase-3 protein were lower in neonatal neutrophils than adult neutrophils, as judged by RT-PCR and Western blot analyses. Moreover, caspase-3 activity was lower in neonatal neutrophils, compared to adult neutrophils. These findings suggest that neonatal neutrophils are intrinsically apoptosis-resistant, which may be due to the low expression of caspase-3.


Asunto(s)
Apoptosis , Caspasa 3/metabolismo , Neutrófilos/citología , Neutrófilos/enzimología , Adulto , Caspasa 3/genética , Regulación Enzimológica de la Expresión Génica , Humanos , Recién Nacido , Persona de Mediana Edad , Adulto Joven
18.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 37(11): 996-1002, 2021 Nov.
Artículo en Zh | MEDLINE | ID: mdl-34809739

RESUMEN

Objective To investigate the effect of insulin-like growth factor 1 (IGF-1) on the migration of alveolar epithelial cells (AECs) and its related mechanisms. Methods The MLE-12 cells (mouse AEC line) were stimulated by IGF-1 and sphingosine 1 phosphate (S1P) in the presence or absence of the PI3K inhibitor Wortmannin. Then, the cell migration was detected by the scratch test and the expression of p-Akt was detected by Western blot. With AECs stimulated by IGF-1, the secretion and expression of S1P were tested by ELISA and Western blot respectively. In the blocking experiment, the effect of IGF-1 on cell migration or p-Akt expression was detected by scratch test or Western blot after the interference of AEC S1P receptor 1 (S1PR1) or the action of S1PR1 blocking antibody. Results After 12 hours of IGF-1 stimulation, the expression of p-Akt in AECs increased and the migration of AECs accelerated. When blocking PI3K signal, the effect of IGF-1 on promoting AEC migration was partially eliminated. IGF-1 induced AECs to produce S1P, which accelerated AEC migration through S1PR1. The expression of p-Akt in AECs increased after S1P stimulation. When blocking the PI3K pathway, the ability of S1P to accelerate the migration of AECs was reduced. When S1PR1 in AECs was blocked or interfered, the effect of IGF-1 on accelerating AEC migration and promoting AEC p-Akt expression was partially reduced. Conclusion IGF-1 activates the PI3K pathway through S1P-S1PR1 signal to promote the migration of AECs.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Esfingosina , Células Epiteliales Alveolares , Animales , Factor I del Crecimiento Similar a la Insulina , Ratones , Proteínas Proto-Oncogénicas c-akt , Receptores de Lisoesfingolípidos/genética , Esfingosina/farmacología
19.
Emerg Microbes Infect ; 10(1): 578-588, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33666534

RESUMEN

Mycobacterium tuberculosis (M. tuberculosis) is the pathogen which causes tuberculosis (TB), a significant human public health threat. Co-infection of M. tuberculosis and the human immunodeficiency virus (HIV), emergence of drug resistant M. tuberculosis, and failure to develop highly effective TB vaccines have limited control of the TB epidemic. Trained immunity is an enhanced innate immune response which functions independently of the adaptive/acquired immune system and responds non-specifically to reinfection with invading agents. Recently, several studies have found trained immunity has the capability to control and eliminate M. tuberculosis infection. Over the past decades, however, the consensus was adaptive immunity is the only protective mechanism by which hosts inhibit M. tuberculosis growth. Furthermore, autophagy plays an essential role in the development of trained immunity. Further investigation of trained immunity, M. tuberculosis infection, and the role of autophagy in this process provide new possibilities for vaccine development. In this review, we present the general characteristics of trained immunity and autophagy. We additionally summarize several examples where initiation of trained immunity contributes to the prevention of M. tuberculosis infection and propose future directions for research in this area.


Asunto(s)
Autofagia , Inmunidad Innata , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/inmunología , Tuberculosis/prevención & control , Inmunidad Adaptativa , Animales , Humanos , Memoria Inmunológica , Vacunación
20.
Blood ; 112(4): 1269-79, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18524989

RESUMEN

Despite the evidence for the role of inflammation in cancer initiation, promotion, and progression, the precise mechanism by which the inflammation within tumor is orchestrated by inflammatory cells remains to be determined. Here, we report that tumor-infiltrating mast cells remodel tumor microenvironment and promote tumor growth. Mast cell infiltration and activation in tumors were mainly mediated by tumor-derived stem cell factor (SCF) and its receptor c-Kit on mast cells. Low concentrations of SCF efficiently induced the chemotactic migration of mast cells. Tumor-infiltrating mast cells, activated by higher concentrations of SCF, expressed multiple proinflammatory factors and increased IL-17 expression in tumors. The activity of NF-kappaB and AP-1 in tumor cells was intensified in the mast cell-remodeled inflammatory microenvironment. SCF-activated mast cells also exacerbated tumor immunosuppression by releasing adenosine and increasing T regulatory cells, which augmented the suppression of T cells and natural killer cells in tumors. These findings emphasize that the remodeling of the tumor microenvironment can actually be initiated by tumor cell-released SCF and suggest that mast cells are not only a participator but also a critical regulator of inflammation and immunosuppression in the tumor microenvironment.


Asunto(s)
Quimiotaxis , Inflamación , Mastocitos/inmunología , Neoplasias/inmunología , Factor de Células Madre/fisiología , Animales , Línea Celular Tumoral , Células Cultivadas , Humanos , Mediadores de Inflamación/análisis , Interleucina-17 , Células Asesinas Naturales/patología , Recuento de Linfocitos , Mastocitos/patología , Ratones , Ratones Endogámicos , FN-kappa B/metabolismo , Trasplante de Neoplasias , Neoplasias/patología , Linfocitos T/patología , Linfocitos T Reguladores/patología , Factor de Transcripción AP-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA