Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 71
1.
Allergol. immunopatol ; 51(4): 31-39, 2023. graf
Article En | IBECS | ID: ibc-222632

Background: Asthma is a common illness with chronic airway inflammation. C1q/tumor necrosis factor (TNF)-related protein 3 (CTRP3) plays a vital role ininflammatory response, but its effect on asthma is imprecise. Herein, we analyzed the functions of CTRP3 in asthma. Methods: The BALB/c mice were randomized into four groups: control, ovalbumin (OVA), OVA+vector, and OVA+CTRP3. The asthmatic mice model was established by OVA stimulation. Overexpression of CTRP3 was implemented by the transfection of corresponding adeno-associated virus 6 (AAV6). The contents of CTRP3, E-cadherin, N-cadherin, smooth muscle alpha-actin (α-SMA), phosphorylated (p)-p65/p65, transforming growth factor-beta 1 (TGFβ1), and p-Smad3/Smad3 were determined by Western blot analysis. The quantity of total cells, eosinophils, neutrophils, and lymphocytes in bronchoalveolar lavage fluid (BALF) was assessed by using a hemocytometer. The contents of tumor necrosis factor-α and interleukin-1β in BALF were examined by enzyme-linked immunesorbent serologic assay. The lung function indicators and airway resistance (AWR) were measured. The bronchial and alveolar structures were evaluated by hematoxylin and eosin staining and sirius red staining. Results: The CTRP3 was downregulated in mice of OVA groups; however, AAV6-CTRP3 treatment markedly upregulated the expression of CTRP3. Upregulation of CTRP3 diminished asthmatic airway inflammation by decreasing the number of inflammatory cells and the contents of proinflammatory factors. CTRP3 markedly lessened AWR and improved lung function in OVA-stimulated mice. Histological analysis found that CTRP3 alleviated OVA-induced airway remodeling in mice. Moreover, CTRP3 modulated NF-κB and TGFβ1/Smad3 pathways in OVA-stimulated mice. Conclusion: CTRP3 alleviated airway inflammation and remodeling in OVA-induced asthmatic mice via regulating NF-κB and TGFβ1/Smad3 pathways (AU)


Animals , Female , Mice , Asthma/immunology , Asthma/metabolism , Inflammation/metabolism , Airway Remodeling , NF-kappa B/immunology , Transforming Growth Factor beta1/immunology , Smad3 Protein/immunology , Disease Models, Animal , Random Allocation , Chronic Disease
2.
Mol Immunol ; 137: 1-10, 2021 09.
Article En | MEDLINE | ID: mdl-34175710

Epidemiological studies have suggested that fine particulate matter (PM2.5) and asthma have been independently associated with pulmonary fibrosis but rarely studied together. Furthermore, it is unknown whether airway fibrosis in asthma is more attributable to water-soluble ions of PM2.5. Our current study was to explore the potential mechanism of PM2.5 and water-soluble components on airway fibrosis in ovalbumin (OVA)-sensitized asthmatic rats. Rats were intratracheally instilled with PM2.5 and water-soluble components every 3 days for 4 times or 8 times. Histopathological examination demonstrated that lung inflammatory and airway fibrosis were induced after PM2.5 and water-soluble components exposure. Meanwhile, PM2.5, in particular water-soluble extracts, increased expression of collagen 1 (COL-1), connective tissue growth factor (CTGF), interleukin-6 (IL-6), transforming growth factor-ß1 (TGF-ß1), Smad family member 3 (Smad3), and p-Smad3, whereas decreased secretion of heme oxygenase-1 (HO-1). However, pretreating asthmatic rats with SB432542, the inhibitor of TGF-ß1, and SIS3 HCl, the antagonist of Smad3, both reversed the activation of airway fibrosis induced by water-soluble extracts. Therefore, TGF-ß1/Smad3 signaling pathway may be responsible for the pathological process of airway fibrosis in asthmatic rats following PM2.5 and water-soluble components exposure.


Lung/immunology , Particulate Matter/immunology , Pulmonary Fibrosis/immunology , Signal Transduction/immunology , Smad3 Protein/immunology , Transforming Growth Factor beta1/immunology , Animals , Asthma/immunology , Disease Models, Animal , Male , Ovalbumin/immunology , Rats , Rats, Sprague-Dawley , Water
3.
Eur J Immunol ; 51(7): 1748-1761, 2021 07.
Article En | MEDLINE | ID: mdl-33811758

Treg are known to have a central role in orchestrating immune responses, but less is known about the destiny of Treg after being activated by specific Ags. This study aimed to investigate the role of superoxide dismutase, an active molecule in the regulation of oxidative stress in the body, in the prevention of Treg apoptosis induced by specific Ags. Ag-specific Tregs were isolated from the DO11.10 mouse intestine. A food allergy mouse model was developed with ovalbumin as the specific Ag and here, we observed that exposure to specific Ag induced Treg apoptosis through converting the precursor of TGF-ß to its mature form inside the Tregs. Oxidative stress was induced in Tregs upon exposure to specific Ags, in which Smad3 bound the latency-associated peptide to induce its degradation, converting the TGF-ß precursor to its mature form, TGF-ß. Suppressing oxidative stress in Tregs alleviated the specific Ag-induced Treg apoptosis in in vitro experiments and suppressed experimental food allergy by preventing the specific Ag-induced Treg apoptosis in the intestine. In conclusion, exposure to specific Ags induces Treg apoptosis and it can be prevented by upregulating superoxide dismutase or suppressing reactive oxidative species in Tregs.


Antigens/immunology , Apoptosis/immunology , Oxidative Stress/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Smad3 Protein/immunology , Superoxide Dismutase/immunology , Transforming Growth Factor beta/immunology , Up-Regulation/immunology
4.
Aging (Albany NY) ; 13(3): 3386-3404, 2021 01 10.
Article En | MEDLINE | ID: mdl-33428605

In this study, we investigated the role of tumor-associated macrophages (TAMs) in the progression of pancreatic ductal adenocarcinoma (PDAC). PDAC patients with higher levels of CD68+ TAMs exhibited shorter overall survival. In Transwell assays, PDAC cells incubated with TAMs or conditioned media from TAM cells (TAM-CM) showed higher migration and invasion rates than controls. PET/CT scan analysis of orthotopic PDAC model mice revealed greater primary tumor growth and liver metastasis in the TAM-CM treatment group than the controls. H&E staining of liver tissues showed significantly higher numbers of metastatic nodules in the TAM-CM treatment group. Heat inactivation of TAM-CM significantly reduced Transwell migration by PDAC cells, suggesting the involvement of one or more secreted proteins in PDAC progression. Transcriptome sequencing analysis of PDAC cells treated with TAM-CM revealed significant enrichment of transforming growth factor-ß (TGF-ß) signaling pathway genes. Western blot and qRT-PCR analysis showed that TAM-CM enhanced PDAC migration cells by inducing epithelial-to-mesenchymal transition through the TGF-ß-Smad2/3/4-Snail signaling axis. The pro-tumorigenic effects of TAMs or TAM-CM were abolished by TGF-ß signaling pathway inhibitors and neutralizing TGF-ß antibody. These results demonstrate that TAMs promote PDAC progression through the TGF-ß signaling pathway.


Carcinoma, Pancreatic Ductal/immunology , Epithelial-Mesenchymal Transition/immunology , Pancreatic Neoplasms/immunology , Tumor-Associated Macrophages/immunology , Animals , Carcinoma, Pancreatic Ductal/secondary , Cell Line, Tumor , Cell Movement , Culture Media, Conditioned , Disease Progression , Gene Expression Profiling , Humans , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Pancreatic Neoplasms/pathology , Prognosis , Smad2 Protein/immunology , Smad3 Protein/immunology , Smad4 Protein/immunology , Snail Family Transcription Factors/immunology , THP-1 Cells , Transforming Growth Factor beta/immunology
5.
Inflamm Res ; 70(1): 109-126, 2021 Jan.
Article En | MEDLINE | ID: mdl-33136175

OBJECTIVE: Cysteinyl leukotrienes (CysLTs), a group of inflammatory lipid mediators, are found elevated in obese-asthmatic patients. Leukotriene D4 (LTD4), a representative CysLT, is implicated in promoting lung inflammation and remodelling in allergic asthma, but its role in non-allergic asthma, especially in obese-asthmatic patients, is not known. Here, using primary human small airway epithelial cells (SAECs) we have investigated the mechanism of LTD4-induced inflammation and remodelling and assessed high proneness of obese mice to develop asthma upon challenge with allergen ovalbumin (OVA). METHODS: Primary human small airway epithelial cells (SAECs) were stimulated with different concentrations of LTD4 for different time intervals and various inflammatory markers were measured through cytokine array, membrane-based ELISA and Western blotting. An air-liquid interface (ALI) model of SAECs was used to study the effects of LTD4-induced remodelling in SAECs using Western blotting, H&E staining and PAS staining. Further, OVA-based murine model was used to examine the propensity of high-fat diet (HFD)-fed obese mice to develop asthma symptoms by studying the infiltration of inflammatory cells (assessed by bronchioalveolar lavage (BAL) cytology) and airway remodelling (assessed by histopathology) upon allergen exposure. RESULTS: The human primary small airway epithelial cells (SAECs) treated with LTD4 showed significant alterations in the levels of inflammatory markers such as GM-CSF, TNF-α, IL-1ß, EGF and eotaxin in dose- and time-dependent manner. Further, LTD4 enhanced the activation of inflammasomes as evidenced by increased levels of NALP3, cleaved caspase-1 and IL-1ß. LTD4 also enhanced inflammation by increasing the expression of COX-2 in SAECs. The airway remodelling markers Vimentin and Muc5AC were found elevated in ALI culture of SAECs when stimulated with LTD4, as it also increased TGF-ß levels and activation of Smad2/3 phosphorylation in SAECs. Last, sensitization and challenge of HFD-fed obese mice with OVA showed increased infiltration of inflammatory cells in BAL and enhanced levels of remodeling phenotypes like loss of cilia, mucus cell metaplasia and collagen deposition in mice lung tissues. CONCLUSION: The results suggest that LTD4 could induce inflammatory response in human airway epithelial cell by activating NALP3 inflammasome. LTD4 could further promote airway epithelial cells' remodelling through TGF-ß/smad2/3-mediated pathway. Our in vivo results suggested that obesity predisposed the OVA challenged mice to develop lung inflammation and remodelling akin to asthma-like phenotypes during obesity.


Airway Remodeling/immunology , Asthma/immunology , Epithelial Cells/immunology , Inflammation/immunology , Leukotriene D4/immunology , Obesity/immunology , Allergens/immunology , Animals , Asthma/pathology , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Cytokines/immunology , Humans , Inflammasomes/immunology , Inflammation/pathology , Leukocyte Count , Male , Mice, Inbred BALB C , Mucin 5AC/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Obesity/pathology , Ovalbumin/immunology , Smad2 Protein/immunology , Smad3 Protein/immunology , Vimentin/immunology
6.
Int J Mol Sci ; 21(19)2020 Sep 28.
Article En | MEDLINE | ID: mdl-32998457

MicroRNAs regulate gene expression of transcriptional factors, which influence Th17/Treg (regulatory T cells) balance, establishing the molecular mechanism of genetic and epigenetic regulation of Treg and Th17 cells is crucial for understanding rheumatoid arthritis (RA) pathogenesis. The study goal was to understand the potential impact of the selected microRNAs expression profiles on Treg/Th17 cells frequency, RA phenotype, the expression profile of selected microRNAs, and their correlation with the expression profiles of selected transcriptional factors: SOCS1, SMAD3, SMAD4, STAT3, STAT5 in RA; we used osteoarthritis (OA) and healthy controls (HCs) as controls. The study was conducted on 14 RA and 11 OA patients, and 15 HCs. Treg/Th17 frequency was established by flow cytometry. Gene expression analysis was estimated by qPCR. We noticed correlations in RA Th17 cells between miR-26 and SMAD3, STAT3, SOCS1; and miR-155 and STAT3-and in RA Treg cells between miR-26 and SOCS1; miR-31, -155 and SMAD3; and miR-155 and SMAD4. In RA Tregs, we found a negative correlation between miR-26, -126 and STAT5a. The expression level of miR-31 in Th17 cells from RA patients with DAS28 ≤ 5.1 is higher and that for miR-24 is greater in Tregs from patients with DAS28 > 5.1. MiR-146a in Tregs is higher in rheumatoid factor (RF) positive RA patients.


Arthritis, Rheumatoid/genetics , MicroRNAs/genetics , Osteoarthritis/genetics , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Adult , Aged , Aged, 80 and over , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Case-Control Studies , Female , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Humans , Male , MicroRNAs/immunology , Middle Aged , Osteoarthritis/immunology , Osteoarthritis/pathology , Phenotype , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Severity of Illness Index , Smad3 Protein/genetics , Smad3 Protein/immunology , Smad4 Protein/genetics , Smad4 Protein/immunology , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology
7.
J Allergy Clin Immunol ; 145(3): 933-946.e4, 2020 03.
Article En | MEDLINE | ID: mdl-31626843

BACKGROUND: Inducible CD4+CD25+ regulatory T (iTreg) cells can become pathogenic effector cells, enhancing lung allergic responses. OBJECTIVE: We aimed to define the underlying cellular and molecular pathways activated by TGF-ß, which determine the suppressor or enhancing activities of iTreg cells. METHODS: Sensitized wild-type and CD8-deficient (CD8-/-) mice were challenged with allergen. Isolated CD4+CD25- T cells were activated by using anti-CD3/anti-CD28. To generate suppressor iTreg cells, cells were then differentiated in the presence of TGF-ß, whereas IL-17-producing effector T cells were additionally exposed to IL-6. After TGF-ß, Smad3 and TGF-ß-activated kinase 1 (TAK1) kinase levels were monitored. The consequences of inhibiting either kinase were determined in vitro and after transfer into CD8-/- recipients. Quantitative PCR and chromatin immunoprecipitation were used to monitor gene expression and histone modifications at the retinoic acid-related orphan receptor γt (Rorγt) locus. RESULTS: In wild-type mice, iTreg cells suppressed lung allergic responses linked to Smad3-dependent forkhead box P3 (Foxp3) expression and IL-10 production. In the presence of IL-6, iTreg cells converted to TH17 cells, mediating a neutrophil-dependent enhancement of lung allergic responses in CD8-/- mice. Conversion was regulated by TAK1. Inhibition or silencing of TAK1 prevented expression of Rorγt and TH17 differentiation through histone modifications of Rorγt; Foxp3 expression and iTreg cell-mediated suppression remained intact. In the same cell, TGF-ß induced coexpression of Smad3 and TAK1 proteins; in the presence of IL-6, expression of Smad3 and Foxp3 but not TAK1 decreased. CONCLUSION: TGF-ß regulates iTreg cell outcomes through 2 distinct signal transduction pathways: one Smad3 dependent and the other TAK1 dependent. The balance of these pathways has important implications in TH17-mediated autoimmune diseases and neutrophil-dependent asthma.


MAP Kinase Kinase Kinases/immunology , Respiratory Hypersensitivity/immunology , Smad3 Protein/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Cell Differentiation/immunology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology
8.
Curr Med Sci ; 39(5): 748-753, 2019 Oct.
Article En | MEDLINE | ID: mdl-31612392

Although several studies confirmed that berberine may attenuate airway inflammation in mice with chronic obstructive pulmonary disease (COPD), its underlying mechanisms were not clear until now. We aimed to establish an experiment mouse model for COPD and to investigate the effects of berberine on airway inflammation and its possible mechanism in COPD model mice induced by cigarette smoke extract (CSE). Twenty SPF C57BL/6 mice were randomly divided into PBS control group, COPD model group, low-dose berberine group and high-dose berberine group, 5 mice in each group. The neutrophils and macrophages were examined by Wright's staining. The levels of inflammatory cytokines TNF-α and IL-6 in bronchoalveolar lavage fluid (BALF) were determined by enzyme-linked immunosorbent assay. The expression levels of TGF-ß1, Smad2 and Smad3 mRNA and proteins in lung tissues were respectively detected by quantitative real-time polymerase chain reaction and Western blotting. It was found that CSE increased the number of inflammation cells in BALF, elevated lung inflammation scores, and enhanced the TGF-ß1/Smads signaling activity in mice. High-dose berberine restrained the alterations in the COPD mice induced by CSE. It was concluded that high-dose berberine ameliorated CSE-induced airway inflammation in COPD mice. TGF-ß1/Smads signaling pathway might be involved in the mechanism. These findings suggested a therapeutic potential of high-dose berberine on the CSE-induced airway inflammation.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Berberine/pharmacology , Lung/drug effects , Pulmonary Disease, Chronic Obstructive/drug therapy , Smad2 Protein/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta1/genetics , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cigarette Smoking/adverse effects , Complex Mixtures/antagonists & inhibitors , Complex Mixtures/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Gene Expression Regulation , Inflammation , Interleukin-6/genetics , Interleukin-6/immunology , Lung/immunology , Lung/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/pathology , Signal Transduction , Smad2 Protein/immunology , Smad3 Protein/immunology , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
9.
Int Immunopharmacol ; 75: 105759, 2019 Oct.
Article En | MEDLINE | ID: mdl-31325729

This study aimed to investigate the effect and underlying mechanism of Methyl helicterilate from Helicteres angustifolia (MHHA) on alcohol-induced hepatic fibrosis. The results showed that MHHA treatment markedly alleviated alcohol-induced liver injury and notably reduced collagen deposition in liver tissue. It significantly enhanced the activity of alcohol dehydrogenase and aldehyde dehydrogenase. Moreover, MHHA treatment markedly decreased the content of inflammatory cytokines, alleviated collagen accumulation, and inhibited the expression of TGF-ß1 and Smad2/3 in liver tissue. The experiments in cells showed that MHHA significantly inhibited HSC activation by blocking TGF-ß1/Smads signaling pathway. Additionally, it notably induced HSC apoptosis by modulating the mitochondria-dependent pathway. The present study demonstrates that MHHA treatment significantly ameliorates alcoholic hepatic fibrosis and the underlying mechanism may be ascribed to the inhibition of the TGF-ß1/Smads pathway and regulation of the mitochondria-mediated apoptotic pathway.


Liver Cirrhosis, Alcoholic/drug therapy , Smad2 Protein/immunology , Smad3 Protein/immunology , Transforming Growth Factor beta1/immunology , Triterpenes/therapeutic use , Animals , Apoptosis/drug effects , Cell Line , Collagen/metabolism , Hepatic Stellate Cells/drug effects , Humans , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/immunology , Liver/pathology , Liver Cirrhosis, Alcoholic/immunology , Liver Cirrhosis, Alcoholic/metabolism , Liver Cirrhosis, Alcoholic/pathology , Male , Rats, Wistar , Signal Transduction/drug effects , Triterpenes/pharmacology
10.
Immunopharmacol Immunotoxicol ; 41(3): 438-445, 2019 Jun.
Article En | MEDLINE | ID: mdl-31119954

Objective: Paeonol is a natural phenolic component isolated from the root bark of peony with multiple pharmacological activities. We investigated the anti-fibrotic effect and underlying mechanism of paeonol. Methods: Twenty-four male C57BL/6J mice were divided into 4 groups (n = 6 in each group), injected with CCl4 to induce liver fibrosis and administrated with paeonol according to the regimen. The serum activity of ALT and AST, and H&E staining were to assess liver injury. Sirius and Masson staining, and hydroxyproline content were to evaluate the degree of liver fibrosis. TNF-α, IL-6, TGF-ß, MDA, GSH-PX, SOD, and CAT were detected to reflect inflammation and oxidative stress. RT-qPCR and Western blot analysis to assess the activation of HSCs and TGF-ß/Smad3 signaling. Results: Paeonol ameliorated liver injury and liver fibrosis, reflected by the decrease of ALT, AST, less lesion in H&E staining, mitigated fibrosis in Sirius and Masson staining, lessened content of hydroxyproline. Paeonol attenuated the level of IL-6 and TNF-α, and elevated the activity of GSH-PX, SOD, and CAT with reducing the level of MDA. The expression of col 1a, α-SMA, vimentin, and desmin were down-regulated and TGF-ß/Smad3 signaling pathway was inhibited. Conclusion: These data demonstrated that paeonol could alleviate CCl4-induced liver fibrosis through suppression of hepatic stellate cells activation via inhibiting the TGF-ß/Smad3 signaling.


Acetophenones/pharmacology , Carbon Tetrachloride Poisoning/drug therapy , Hepatic Stellate Cells/immunology , Liver Cirrhosis/drug therapy , Signal Transduction/drug effects , Smad3 Protein/immunology , Transforming Growth Factor beta/immunology , Animals , Carbon Tetrachloride Poisoning/immunology , Carbon Tetrachloride Poisoning/pathology , Hepatic Stellate Cells/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Male , Mice , Signal Transduction/immunology
11.
Biochim Biophys Acta Mol Basis Dis ; 1865(7): 1772-1781, 2019 07 01.
Article En | MEDLINE | ID: mdl-29940270

Macrophages exhibit heterogeneity and plasticity and imbalance between pro-inflammatory and anti-inflammatory macrophages plays a critical role in atherosclerosis progression. Telomerase reverse transcriptase (TERT) in macrophages can be activated by nuclear factor-kappa B (NF-κB), but the regulation of telomerase activation on macrophages polarization remains unknown. We previously identified microRNA-216a (miR-216a) to promote inflammation through directly targeting the Smad3/NF-κB pathway. The present study aimed to assess whether miR-216a can regulate telomerase activity and promote macrophages polarization during atherosclerosis progression. The results verified that TERT was highly expressed in macrophages of human carotid atherosclerotic plaques. miR-216a was found to promote telomerase activation in macrophages by 4.5-fold (P = 0.002) through the Smad3/NF-κB pathway. miR-216a also induced macrophages senescence characterized by senescence-associated-ß-galactosidase activity and p53 and p16 expression. TERT overexpression promoted the transformation of M2 to M1 while this conversion was suppressed once TERT was inhibited, and the related inflammatory factors and lipid uptake ability of M1 cells were also increased by TERT. In the carotid atherosclerotic plaques from miR-216a-treated apolipoprotein E-/- mice, the numbers of M1 macrophages were increased whereas M2 cells reduced, accompanying with inhibited Smad3 expression and upregulated inflammatory markers and TERT activity. Furthermore, plasma miR-216a level was specifically higher in patients with vulnerable mixed plaques (n = 181) than those with calcified plaques (n = 73) and controls (n = 264). In summary, our findings first revealed a new molecular mechanism of macrophage polarization involving telomerase activation induced by miR-216a through the Smad3/NF-κB signaling, which might serve as a potential therapeutic target for atherosclerosis progression.


Atherosclerosis/genetics , MicroRNAs/genetics , NF-kappa B/immunology , Smad3 Protein/immunology , Telomerase/genetics , Animals , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Line , Disease Progression , Enzyme Activation , Humans , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Telomerase/immunology , Up-Regulation
12.
Biomed Res Int ; 2018: 9064073, 2018.
Article En | MEDLINE | ID: mdl-30364052

CD4+CD25+Foxp3+ Tregs control the immune response and maintain immune homeostasis. This study examined whether Tregs can affect mouse enteritis and the Foxp3 (Forkhead transcription factor) transcriptional pathway. Mouse CD4+CD25+ Treg cells were labelled using CFSE (5,6-carboxyfluorescein diacetate succinimidyl ester) and transferred to enteritis model mice. The mice were randomly divided into an enteritis group, a Treg-infusion group, a Treg-inhibiting group, and a control group. Histopathology, ELISA, flow cytometry, western blot, immunohistochemistry, and immunofluorescence were performed. Our results demonstrated that CD4+CD25+ Tregs were successfully transferred. The disease activity index (DAI) scores in the Tregs-infusion group were lower than those of the enteritis and Tregs-inhibiting groups. The number of goblet cells and inflammatory cells was reduced, and the levels of IL-1ß, TNF-α, NO, and PGE2 were significantly decreased in the Tregs-infusion group compared to those in the enteritis group (p<0.05). The number of CD4+CD25+Foxp3+ Tregs and CD4+IL-17A+ Th17 cells in the mesenteric lymph nodes differed significantly from the enteritis and Tregs-inhibiting groups (p<0.05). There were more Foxp3+ Tregs and Smad3 and NFAT2 infiltrated into the duodenum after adoptive transfer of CD4+CD25+ Tregs, which was a significant difference relative to the enteritis group (p<0.05). This study demonstrated that adoptive transfer of CD4+CD25+ Tregs can decrease mouse enteritis. Foxp3 expression may be improved through the Smad3 and NFAT2 signalling pathways.


Adoptive Transfer , Enteritis/therapy , Forkhead Transcription Factors/immunology , Gene Expression Regulation/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/transplantation , Animals , Disease Models, Animal , Enteritis/immunology , Enteritis/pathology , Male , Mice , Mice, Inbred BALB C , NFATC Transcription Factors/immunology , Smad3 Protein/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/immunology , Th17 Cells/pathology
13.
Cancer Immunol Res ; 6(8): 965-977, 2018 08.
Article En | MEDLINE | ID: mdl-29915022

Natural killer (NK) cells, early effectors in anticancer immunity, are paralyzed by TGFß1, an immunosuppressive cytokine produced by cancer cells. Development and activity of NK cells are largely inhibited in the Smad3-dependent tumor microenvironment. Here, we used genetic engineering to generate a stable SMAD3-silencing human NK cell line, NK-92-S3KD, whose cancer-killing activity and cytokine production were significantly enhanced under TGFß1-rich condition compared with the parental cell line. Interestingly, we identified that the IFNG gene is a direct E4BP4 target gene. Thus, silencing of SMAD3 allows upregulation of E4BP4 that subsequently promoting interferon-γ (IFNγ) production in the NK-92-S3KD cells. More importantly, NK-92-S3KD immunotherapy increases the production of not only IFNγ, but also granzyme B and perforin in tumors; therefore, inhibiting cancer progression in two xenograft mouse models with human hepatoma (HepG2) and melanoma (A375). Thus, the NK-92-S3KD cell line may be useful for the clinical immunotherapy of cancer. Cancer Immunol Res; 6(8); 965-77. ©2018 AACR.


Immunotherapy/methods , Killer Cells, Natural/immunology , Liver Neoplasms, Experimental/therapy , Melanoma, Experimental/therapy , Smad3 Protein/genetics , Animals , Cell Line , Extracellular Matrix Proteins/immunology , Gene Silencing , Genetic Engineering/methods , Humans , Interferon-gamma/biosynthesis , Liver Neoplasms, Experimental/immunology , Melanoma, Experimental/immunology , Mice, Inbred NOD , Mice, Knockout , Smad3 Protein/immunology , Transforming Growth Factor beta/immunology , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
14.
Mol Med Rep ; 18(2): 2088-2096, 2018 Aug.
Article En | MEDLINE | ID: mdl-29916550

The present study aimed to investigate the effect of epigallocatechin gallate (EGCG) on airway inflammation in mice with bronchial asthma, and the regulatory mechanism of transforming growth factor (TGF)­ß1 signaling pathway, so as to provide theoretical basis for research and development of a novel drug for clinical treatment. Mouse models of bronchial asthma were established and injected with dexamethasone and EGCG via the caudal vein. 7 days later, bronchoalveolar tissue was collected for hematoxylin and eosin staining. Determination of airway resistance (AWR) and lung function in mice was detected. Serum was separated for cytometric bead array to detect the changes in inflammatory factors. Bronchoalveolar lavage fluid was collected for eosinophil and neutrophil counts. Fresh blood was obtained for flow cytometry to determine the percentages of Th17 and Treg cells. Bronchovesicular tissue was utilized for western blot assay and reverse transcription­quantitative polymerase chain reaction to determine the proteins and transcription factors in the TGF­ß1 pathway. EGCG 20 mg/kg significantly reduced asthmatic symptoms, lung inflammatory cell infiltration, and the inflammatory factor levels of interleukin (IL)­2, IL­6 and tumor necrosis factor (TNF)­α. In addition, it increased the levels of inflammatory factors, including IL­10, diminished the percentage of Th17 cells, increased the percentage of Treg cells, and decreased the expression of TGF­ß1 and phosphorylated (p)­Smad2/3 expression. Following the inhibition of the TGF­ß1 receptor, the anti­inflammatory effect of EGCG disappeared, and the expression of TGF­ß1 and p­Smad2/3 increased. EGCG attenuated airway inflammation in asthmatic mice, decreased the percentage of Th17 cells and increased the percentage of Treg cells. The anti­inflammatory effect of EGCG is achieved via the TGF­ß1 signaling pathway.


Asthma/drug therapy , Catechin/analogs & derivatives , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Transforming Growth Factor beta1/immunology , Animals , Asthma/immunology , Asthma/pathology , Catechin/pharmacology , Cytokines/immunology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Male , Mice , Mice, Inbred BALB C , Signal Transduction/immunology , Smad2 Protein/immunology , Smad3 Protein/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology
15.
J Neuroimmunol ; 314: 30-41, 2018 01 15.
Article En | MEDLINE | ID: mdl-29169800

Depending on the duration and severity, psychological tension and physical stress can enhance or suppress the immune system in both humans and animals. Although it has been established that chronic stress exerts a significant suppressive effect on immune function, the mechanisms by which affects immune responses remain elusive. By employing an in vivo murine system, we revealed that TGF-ß1/Smad2/3/Foxp3 axis was remarkably activated following chronic stress. Furthermore, TLR9 and p38 MAPK played a critical role in the activation of TGF-ß1/Smad2/3/Foxp3 signaling cascade. Moreover, inhibition of TGF-ß1/Smad2/3/Foxp3 or p38 significantly attenuated chronic stress-induced lymphocyte apoptosis and apoptosis-related proteins, as well as the differentiation of T regulatory cells in spleen. Interestingly, disequilibrium of pro-inflammatory and anti-inflammatory cytokines balance caused by chronic stress was also rescued by blocking TGF-ß1/Smad2/3/Foxp3 axis. These findings yield insight into a novel mechanism by which chronic stress modulates immune functions and identifies new targets for the development of novel anti-immune suppressant medications.


Immune Tolerance/immunology , Signal Transduction/immunology , Stress, Psychological/immunology , Animals , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Male , Mice , Mice, Inbred BALB C , Smad2 Protein/immunology , Smad2 Protein/metabolism , Smad3 Protein/immunology , Smad3 Protein/metabolism , Stress, Psychological/metabolism , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism
16.
Patol Fiziol Eksp Ter ; 61(2): 4-9, 2017.
Article Ru | MEDLINE | ID: mdl-29215829

Objective. Reprogramming of M1 macrophage phenotype with inhibited M2 phenotype transcription factors, such as STAT3, STAT6 and SMAD and assess their impact on the development of Ehrlich carcinoma (EC) in vitro and in vivo. Methods. Tumor growth in vitro was initiated by addition of EC cells in RPMI-1640 culture medium and in vivo by intraperitoneal of EC cell injection into mice. Results. It was found that M1-STAT3/6- SMAD3 macrophages have a pronounced anti-tumor effect in vitro, and in vivo, which was greater than anti-tumor effects of M1, M1-STAT 3/6, M1-SMAD3 macrophages and cisplatin. Conclusion. M1 macrophages with inhibited STAT3, STAT6 and/or SMAD3 effectively restrict tumor growth. The findings justify the development of new anti-tumor cell therapy technology.


Carcinoma, Ehrlich Tumor , Cellular Reprogramming/immunology , Macrophages/immunology , STAT3 Transcription Factor/immunology , STAT6 Transcription Factor/immunology , Smad3 Protein/immunology , Animals , Carcinoma, Ehrlich Tumor/immunology , Carcinoma, Ehrlich Tumor/pathology , Carcinoma, Ehrlich Tumor/therapy , Mice , Mice, Inbred BALB C
17.
HLA ; 90(6): 329-334, 2017 12.
Article En | MEDLINE | ID: mdl-29106067

Genetic studies in complex diseases have been highly successful, but have also been largely one-dimensional: predominantly focusing on the genetic contribution to disease susceptibility. While this is undoubtedly important-indeed it is a pre-requisite for understanding the mechanisms underlying disease development-there are many other important aspects of disease biology that have received comparatively little attention. In this review, I will discuss how existing genetic data can be leveraged to provide new insights into other aspects of disease biology, why such insights could change the way we think about complex disease, and how this could provide opportunities for better therapies and/or facilitate personalised medicine. To do this, I will use the example of Crohn's disease-a chronic form of inflammatory bowel disease that has been one of the main success stories in complex disease genetics. Indeed, thanks to genetic studies, we now have a much more detailed understanding of the processes involved in Crohn's disease development, but still know relatively little about what determines the subsequent disease course (prognosis) and why this differs so considerably between individuals. I will discuss how we came to realise that genetic variation plays an important role in determining disease prognosis and how this has changed the way we think about Crohn's disease genetics. This will illustrate how phenotypic data can be used to leverage new insights from genetic data and will provide a broadly applicable framework that could yield new insights into the biology of multiple diseases.


Crohn Disease/genetics , Gene Expression Regulation , Genetic Predisposition to Disease , Genetic Variation , Genome, Human , Alleles , Crohn Disease/diagnosis , Crohn Disease/immunology , Crohn Disease/pathology , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/immunology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Genetic Loci , Genome-Wide Association Study , Humans , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/immunology , Prognosis , Severity of Illness Index , Smad3 Protein/genetics , Smad3 Protein/immunology , Toll-Like Receptor 10/genetics , Toll-Like Receptor 10/immunology
18.
Cell Immunol ; 319: 43-52, 2017 Sep.
Article En | MEDLINE | ID: mdl-28864263

Cholera toxin B subunit fusion to autoantigens such as proinsulin (CTB-INS) down regulate dendritic cell (DC) activation and stimulate synthesis of DC immunosuppressive cytokines. Recent studies of CTB-INS induction of immune tolerance in human DCs indicate that increased biosynthesis of indoleamine 2,3-dioxygenase (IDO1) may play an important role in CTB-INS vaccine suppression of DC activation. Studies in murine models suggest a role for transforming growth factor beta (TGF-ß) in the stimulation of IDO1 biosynthesis, for the induction of tolerance in DCs. Here, we investigated the contribution of TGF-ß superfamily proteins to CTB-INS induction of IDO1 biosynthesis in human monocyte-derived DCs (moDCs). We show that CTB-INS upregulates the level of TGF-ß1, activin-A and the TGF-ß activator, integrin αvß8 in human DCs. However, inhibition of endogenous TGF-ß, activin-A or addition of biologically active TGF-ß1, and activin-A, did not inhibit or stimulate IDO1 biosynthesis in human DCs treated with CTB-INS. While inhibition with the kinase inhibitor, RepSox, blocked SMAD2/3 phosphorylation and diminished IDO1 biosynthesis in a concentration dependent manner. Specific blocking of the TGF-ß type 1 kinase receptor with SB-431542 did not arrest IDO1 biosynthesis, suggesting the involvement of a different kinase pathway other than TGF-ß type 1 receptor kinase in CTB-INS induction of IDO1 in human moDCs. Together, our experimental findings identify additional immunoregulatory proteins induced by the CTB-INS fusion protein, suggesting CTB-INS may utilize multiple mechanisms in the induction of tolerance in human moDCs.


Dendritic Cells/drug effects , Gene Expression Regulation/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Recombinant Fusion Proteins/pharmacology , Transforming Growth Factor beta1/genetics , Activins/genetics , Activins/immunology , Animals , Cell Differentiation , Cholera Toxin/genetics , Cholera Toxin/immunology , Cloning, Molecular , Dendritic Cells/cytology , Dendritic Cells/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Integrins/genetics , Integrins/immunology , Mice , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Primary Cell Culture , Proinsulin/genetics , Proinsulin/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/immunology , Smad3 Protein/genetics , Smad3 Protein/immunology , Transforming Growth Factor beta1/immunology
19.
J Cell Mol Med ; 21(12): 3264-3276, 2017 Dec.
Article En | MEDLINE | ID: mdl-28609013

Radiation-induced lung injury (RILI) is one of the most common and fatal complications of thoracic radiotherapy. It is characterized with two main features including early radiation pneumonitis and fibrosis in later phase. This study was to investigate the potential radioprotective effects of polydatin (PD), which was shown to exert anti-inflammation and anti-oxidative capacities in other diseases. In this study, we demonstrated that PD-mitigated acute inflammation and late fibrosis caused by irradiation. PD treatment inhibited TGF-ß1-Smad3 signalling pathway and epithelial-mesenchymal transition. Moreover, radiation-induced imbalance of Th1/Th2 was also alleviated by PD treatment. Besides its free radical scavenging capacity, PD induced a huge increase of Sirt3 in culture cells and lung tissues. The level of Nrf2 and PGC1α in lung tissues was also elevated. In conclusion, our data showed that PD attenuated radiation-induced lung injury through inhibiting epithelial-mesenchymal transition and increased the expression of Sirt3, suggesting PD as a novel potential radioprotector for RILI.


Acute Lung Injury/prevention & control , Epithelial-Mesenchymal Transition/drug effects , Glucosides/pharmacology , Radiation Pneumonitis/prevention & control , Radiation-Protective Agents/pharmacology , Sirtuin 3/genetics , Stilbenes/pharmacology , Acute Lung Injury/genetics , Acute Lung Injury/immunology , Acute Lung Injury/pathology , Animals , Cell Line , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/radiation effects , Epithelial-Mesenchymal Transition/radiation effects , Female , Gene Expression Regulation , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung/radiation effects , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/immunology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/immunology , Radiation Pneumonitis/genetics , Radiation Pneumonitis/immunology , Radiation Pneumonitis/pathology , Signal Transduction , Sirtuin 3/immunology , Smad3 Protein/genetics , Smad3 Protein/immunology , Th1-Th2 Balance/drug effects , Th1-Th2 Balance/radiation effects , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology
20.
Nat Commun ; 8: 15818, 2017 06 09.
Article En | MEDLINE | ID: mdl-28598443

Regulatory T (Treg) cells are important for the maintenance of immune homoeostasis and prevention of autoimmune diseases. Epigenetic modifications have been reported to modulate autoimmunity by altering Treg cell fate. Here we show that the H3K4 methyltransferase Ash1l facilitates TGF-ß-induced Treg cell polarization in vitro and protects mice from T cell-mediated colitis in vivo. Ash1l upregulates Smad3 expression by directly targeting Smad3 promoter to increase local H3K4 trimethylation. Furthermore, we identify an lncRNA, namely lnc-Smad3, which interacts with the histone deacetylase HDAC1 and silences Smad3 transcription. After TGF-ß stimulation, activated Smad3 suppresses lnc-Smad3 transcription, thereby recovering the Smad3 promoter accessibility to Ash1l. By revealing the opposite regulatory functions of Ash1l and lnc-Smad3 in Smad3 expression, our data provide insights for the epigenetic control of Treg cell fate to potentially aid in the development of therapeutic intervention for autoimmune diseases.


Autoimmunity , Cell Polarity , Histone-Lysine N-Methyltransferase/immunology , RNA, Long Noncoding/genetics , Smad3 Protein/genetics , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes/immunology , Adult , Aged , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Epigenesis, Genetic , Gene Expression Regulation , Gene Silencing , Histone Deacetylase 1/genetics , Histone Deacetylase 1/immunology , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/immunology , Humans , Methylation , Mice , Mice, Inbred C57BL , Middle Aged , Promoter Regions, Genetic , RNA, Long Noncoding/immunology , Smad3 Protein/immunology , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Transforming Growth Factor beta/immunology
...