Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Aging (Albany NY) ; 13(3): 3386-3404, 2021 01 10.
Article in English | MEDLINE | ID: mdl-33428605

ABSTRACT

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.


Subject(s)
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
2.
Inflamm Res ; 70(1): 109-126, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33136175

ABSTRACT

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.


Subject(s)
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
3.
Curr Med Sci ; 39(5): 748-753, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31612392

ABSTRACT

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.


Subject(s)
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
4.
Int Immunopharmacol ; 75: 105759, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31325729

ABSTRACT

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.


Subject(s)
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
5.
Proc Natl Acad Sci U S A ; 116(3): 982-987, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30593560

ABSTRACT

Phosphatase PP2A expression levels are positively correlated to the clinical severity of systemic lupus erythematosus (SLE) and IL17A cytokine overproduction, indicating a potential role of PP2A in controlling TH17 differentiation and inflammation. By generating a mouse strain with ablation of the catalytic subunit α of PP2A in peripheral mature T cells (PP2A cKO), we demonstrate that the PP2A complex is essential for TH17 differentiation. These PP2A cKO mice had reduced TH17 cell numbers and less severe disease in an experimental autoimmune encephalomyelitis (EAE) model. PP2A deficiency also ablated C-terminal phosphorylation of SMAD2 but increased C-terminal phosphorylation of SMAD3. By regulating the activity of RORγt via binding, the changes in the phosphorylation status of these R-SMADs reduced Il17a gene transcription. Finally, PP2A inhibitors showed similar effects on TH17 cells as were observed in PP2A cKO mice, i.e., decreased TH17 differentiation and relative protection of mice from EAE. Taken together, these data demonstrate that phosphatase PP2A is essential for TH17 differentiation and that inhibition of PP2A could be a possible therapeutic approach to controlling TH17-driven autoimmune diseases.


Subject(s)
Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental , Protein Phosphatase 2 , Th17 Cells/immunology , Transcription, Genetic/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Interleukin-17/genetics , Interleukin-17/immunology , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Phosphorylation/genetics , Phosphorylation/immunology , Protein Phosphatase 2/genetics , Protein Phosphatase 2/immunology , Smad2 Protein/genetics , Smad2 Protein/immunology , Th17 Cells/pathology
6.
Mol Med Rep ; 18(2): 2088-2096, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29916550

ABSTRACT

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.


Subject(s)
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
7.
J Neuroimmunol ; 314: 30-41, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29169800

ABSTRACT

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.


Subject(s)
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
8.
Nat Commun ; 8(1): 1917, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29203869

ABSTRACT

The melastatin-like transient-receptor-potential-7 protein (TRPM7), harbouring a cation channel and a serine/threonine kinase, has been implicated in thymopoiesis and cytokine expression. Here we show, by analysing TRPM7 kinase-dead mutant (Trpm7 R/R ) mice, that the enzymatic activity of the receptor is not essential for thymopoiesis, but is required for CD103 transcription and gut-homing of intra-epithelial lymphocytes. Defective T cell gut colonization reduces MHCII expression in intestinal epithelial cells. Mechanistically, TRPM7 kinase activity controls TGF-ß-induced CD103 expression and pro-inflammatory T helper 17, but not regulatory T, cell differentiation by modulating SMAD2. Notably, we find that the TRPM7 kinase activity promotes gut colonization by alloreactive T cells in acute graft-versus-host disease. Thus, our results unravel a function of TRPM7 kinase in T cell activity and suggest a therapeutic potential of kinase inhibitors in averting acute graft-versus-host disease.


Subject(s)
Graft vs Host Disease/genetics , Intestines/immunology , Lymphopoiesis/genetics , T-Lymphocytes, Regulatory/cytology , TRPM Cation Channels/genetics , Th17 Cells/cytology , Animals , Antigens, CD/immunology , Cell Differentiation/genetics , Genes, MHC Class II/genetics , Genes, MHC Class II/immunology , Graft vs Host Disease/immunology , Integrin alpha Chains/immunology , Mice , Mutation , Smad2 Protein/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , TRPM Cation Channels/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/immunology
9.
Cell Immunol ; 319: 43-52, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28864263

ABSTRACT

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.


Subject(s)
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
10.
Arthritis Res Ther ; 19(1): 137, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28615031

ABSTRACT

BACKGROUND: Leucine-rich alpha 2 glycoprotein (LRG) has been identified as a serum protein elevated in patients with active rheumatoid arthritis (RA). Although the function of LRG is ill-defined, LRG binds with transforming growth factor (TGF)-ß and enhances Smad2 phosphorylation. Considering that the imbalance between T helper 17 (Th17) cells and regulatory T cells (Treg) plays important roles in the pathogenesis of RA, LRG may affect arthritic pathology by enhancing the TGF-ß-Smad2 pathway that is pivotal for both Treg and Th17 differentiation. The purpose of this study was to explore the contribution of LRG to the pathogenesis of arthritis, with a focus on the role of LRG in T cell differentiation. METHODS: The differentiation of CD4 T cells and the development of collagen-induced arthritis (CIA) were examined in wild-type mice and LRG knockout (KO) mice. To examine the influence of LRG on T cell differentiation, naïve CD4 T cells were isolated from LRG KO mice and cultured under Treg- or Th17-polarization condition in the absence or presence of recombinant LRG. RESULTS: In the CIA model, LRG deficiency led to ameliorated arthritis and reduced Th17 differentiation with no influence on Treg differentiation. By addition of recombinant LRG, the expression of IL-6 receptor (IL-6R) was enhanced through TGF-ß-Smad2 signaling. In LRG KO mice, the IL-6R expression and IL-6-STAT3 signaling was attenuated in naïve CD4 T cells, compared to wild-type mice. CONCLUSIONS: Our findings suggest that LRG upregulates IL-6R expression in naïve CD4 T cells by the enhancement of TGF-ß-smad2 pathway and promote Th17 differentiation and arthritis development.


Subject(s)
Arthritis, Experimental/immunology , Cell Differentiation/immunology , Glycoproteins/immunology , Signal Transduction , T-Lymphocytes, Helper-Inducer/immunology , Th17 Cells/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-6 , Signal Transduction/immunology , Smad2 Protein/immunology , Transforming Growth Factor beta/immunology
11.
J Immunol ; 197(11): 4344-4350, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27793996

ABSTRACT

Immune tolerance between the fetus and mother represents an active process by which the developing fetus must not mount immune responses to noninherited Ags on chimeric maternal cells that reside in fetal tissue. This is, in part, mediated by the suppressive influence of CD4+FOXP3+CD25+ regulatory T cells (Tregs). Fetal secondary lymphoid organs have an increased frequency of Tregs and, as compared with adult T cells, fetal naive CD4+ T cells exhibit a strong predisposition to differentiate into Tregs when stimulated. This effect is mediated by the TCR and TGF-ß pathways, and fetal T cells show significantly increased Treg differentiation in response to anti-CD3 and TGF-ß stimulation. Naive fetal T cells also exhibit increased signaling through the TGF-ß pathway, with these cells demonstrating increased expression of the signaling mediators TGF-ßRI, TGF-ßRIII, and SMAD2, and higher levels of SMAD2/SMAD3 phosphorylation. Increased fetal Treg differentiation is mediated by the RNA-binding protein Lin28b, which is overexpressed in fetal T cells as compared with adult cells. When Lin28b expression is decreased in naive fetal T cells, they exhibit decreased Treg differentiation that is associated with decreased TGF-ß signaling and lowered expression of TGF-ßRI, TGF-ßRIII, and SMAD2. Lin28b regulates the maturation of let-7 microRNAs, and these TGF-ß signaling mediators are let-7 targets. We hypothesize that loss of Lin28b expression in fetal T cells leads to increased mature let-7, which causes decreased expression of TGF-ßRI, TGF-ßRIII, and SMAD2 proteins. A reduction in TGF-ß signaling leads to reduced Treg numbers.


Subject(s)
Cell Differentiation/immunology , Fetus/immunology , RNA-Binding Proteins/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , CD3 Complex/immunology , Female , Fetus/cytology , Gene Expression Regulation, Developmental/immunology , Humans , Male , MicroRNAs/immunology , Protein Serine-Threonine Kinases/immunology , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Antigen, T-Cell/immunology , Receptors, Transforming Growth Factor beta/immunology , Smad2 Protein/immunology , T-Lymphocytes, Regulatory/cytology
12.
Cell Stress Chaperones ; 21(2): 239-49, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26577463

ABSTRACT

Pulmonary fibrosis (PF) can severely disrupt lung function, leading to fatal consequences. Salidroside is a principal active ingredient of Rhodiola rosea and has recently been reported to protect against lung injures. The present study was aimed at exploring its therapeutic effects on PF. Lung fibrotic injuries were induced in SD rats by a single intratracheal instillation of 5 mg/kg bleomycin (BLM). Then, these rats were administrated with 50, 100, or 200 mg/kg salidroside for 28 days. BLM-triggered structure distortion, collagen overproduction, excessive inflammatory infiltration, and pro-inflammatory cytokine release, and oxidative stress damages in lung tissues were attenuated by salidroside in a dose-dependent manner. Furthermore, salidroside was noted to inhibit IκBα phosphorylation and nuclear factor kappa B (NF-κB) p65 nuclear accumulation while activating Nrf2-antioxidant signaling in BLM-treated lungs. Downregulation of E-cadherin and upregulation of vimentin, fibronectin, and α-smooth muscle actin (α-SMA) indicated an epithelial-mesenchymal transition (EMT)-like shift in BLM-treated lungs. These changes were suppressed by salidroside. The expression of TGF-ß1 and the phosphorylation of its downstream targets, Smad-2/-3, were enhanced by BLM, but weakened by salidroside. Additionally, salidroside was capable of reversing the recombinant TGF-ß1-induced EMT-like changes in alveolar epithelial cells in vitro. Our study reveals that salidroside's protective effects against fibrotic lung injuries are correlated to its anti-inflammatory, antioxidative, and antifibrotic properties.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Glucosides/therapeutic use , Lung/drug effects , Phenols/therapeutic use , Pulmonary Fibrosis/drug therapy , Signal Transduction/drug effects , Animals , Anti-Inflammatory Agents/chemistry , Antioxidants/chemistry , Bleomycin , Cell Line , Glucosides/chemistry , Humans , Lung/immunology , Lung/pathology , NF-E2-Related Factor 2/immunology , NF-kappa B/immunology , Phenols/chemistry , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Rats, Sprague-Dawley , Rhodiola/chemistry , Smad2 Protein/immunology , Smad3 Protein/immunology , Transforming Growth Factor beta1/immunology
13.
Int Immunopharmacol ; 28(1): 707-14, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26258626

ABSTRACT

Apoptosis of alveolar epithelial cells has been implicated in the pathogenesis of acute lung injury. Phospholipid transfer protein (PLTP) may play a role in apoptosis. In the present study, the effect of the novel function of PLTP in cigarette smoke extract (CSE)-induced apoptosis of alveolar epithelial cells and the possible mechanism were examined. Male Wistar rats were exposed to air and cigarette smoke (n=10/exposure) for 6h/day on 3 consecutive days, then the lungs were sectioned and examined. To investigate effects on alveolar epithelial cells, rat alveolar epithelial cells (RLE-6TN) were treated with different concentrations of CSE for various times. siRNA for PLTP was transfected into cells and an inhibitor of the transforming growth factor-ß1 (TGF-ß1) type I receptor was administered prior to CSE exposure. Apoptosis was measured, and mRNA expression of PLTP and TGF-ß1 and protein levels of PLTP, TGF-ß1, p-Smad2 and cleaved caspase-3 were analyzed. The results showed that apoptosis, as well as expression of PLTP, TGF-ß1, p-Smad2 and cleaved caspase-3 were all significantly increased after CSE stimulation (P<0.05). Furthermore, the expression of TGF-ß1, p-Smad2 and cleaved caspase-3 induced by CSE could be partly abrogated by knockdown of PLTP. The expression of PLTP showed no significant change as a result of TGF-ß1 receptor inhibition, while cleaved caspase-3 showed a remarkable reduction. PLTP may act as an upstream signal molecule of the TGF-ß1/Smad2 pathway and is likely to be involved in CSE-induced apoptosis of alveolar epithelial cells.


Subject(s)
Apoptosis/drug effects , Phospholipid Transfer Proteins/metabolism , Pulmonary Alveoli/drug effects , Smad2 Protein/metabolism , Smoking/adverse effects , Tobacco Products/toxicity , Transforming Growth Factor beta1/metabolism , Animals , Blotting, Western , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Inhalation Exposure , Lung Injury/chemically induced , Lung Injury/immunology , Lung Injury/pathology , Male , Phospholipid Transfer Proteins/immunology , Pulmonary Alveoli/immunology , Pulmonary Alveoli/pathology , Rats, Wistar , Real-Time Polymerase Chain Reaction , Signal Transduction , Smad2 Protein/immunology , Smoke , Transforming Growth Factor beta1/immunology
14.
Am J Respir Crit Care Med ; 192(8): 998-1008, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26192556

ABSTRACT

RATIONALE: The etiology of schistosomiasis-associated pulmonary arterial hypertension (PAH), a major cause of PAH worldwide, is poorly understood. Schistosoma mansoni exposure results in prototypical type-2 inflammation. Furthermore, transforming growth factor (TGF)-ß signaling is required for experimental pulmonary hypertension (PH) caused by Schistosoma exposure. OBJECTIVES: We hypothesized type-2 inflammation driven by IL-4 and IL-13 is necessary for Schistosoma-induced TGF-ß-dependent vascular remodeling. METHODS: Wild-type, IL-4(-/-), IL-13(-/-), and IL-4(-/-)IL-13(-/-) mice (C57BL6/J background) were intraperitoneally sensitized and intravenously challenged with S. mansoni eggs to induce experimental PH. Right ventricular catheterization was then performed, followed by quantitative analysis of the lung tissue. Lung tissue from patients with schistosomiasis-associated and connective tissue disease-associated PAH was also systematically analyzed. MEASUREMENTS AND MAIN RESULTS: Mice with experimental Schistosoma-induced PH had evidence of increased IL-4 and IL-13 signaling. IL-4(-/-)IL-13(-/-) mice, but not single knockout IL-4(-/-) or IL-13(-/-) mice, were protected from Schistosoma-induced PH, with decreased right ventricular pressures, pulmonary vascular remodeling, and right ventricular hypertrophy. IL-4(-/-)IL-13(-/-) mice had less pulmonary vascular phospho-signal transducer and activator of transcription 6 (STAT6) and phospho-Smad2/3 activity, potentially caused by decreased TGF-ß activation by macrophages. In vivo treatment with a STAT6 inhibitor and IL-4(-/-)IL-13(-/-) bone marrow transplantation also protected against Schistosoma-PH. Lung tissue from patients with schistosomiasis-associated and connective tissue disease-associated PAH had evidence of type-2 inflammation. CONCLUSIONS: Combined IL-4 and IL-13 deficiency is required for protection against TGF-ß-induced pulmonary vascular disease after Schistosoma exposure, and targeted inhibition of this pathway is a potential novel therapeutic approach for patients with schistosomiasis-associated PAH.


Subject(s)
Hypertension, Pulmonary/immunology , Interleukin-13/immunology , Interleukin-4/immunology , Macrophages/immunology , Schistosomiasis mansoni/immunology , Animals , Bone Marrow Transplantation , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Humans , Hypertension, Pulmonary/etiology , Inflammation , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-13/genetics , Interleukin-4/genetics , Interleukin-4 Receptor alpha Subunit/immunology , Interleukin-4 Receptor alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , STAT6 Transcription Factor/immunology , STAT6 Transcription Factor/metabolism , Schistosoma mansoni , Schistosomiasis mansoni/complications , Smad2 Protein/immunology , Smad2 Protein/metabolism , Smad3 Protein/immunology , Smad3 Protein/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/immunology , Vascular Remodeling
15.
J Leukoc Biol ; 98(5): 703-12, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26153417

ABSTRACT

DGK-ζ is a negative regulator of TCR signaling that causes degradation of the second messenger DAG, terminating DAG-mediated activation of Ras and PKCθ. Cytotoxic T cells deficient in DGK-ζ demonstrate enhanced effector functions in vitro and antitumor activity in vivo, perhaps because of insensitivity to inhibitory cytokines. We sought to determine whether the enhanced responsiveness of DGK-ζ-deficient T cells renders them insensitive to the inhibitory cytokine TGF-ß and to determine how the loss of DGK-ζ facilitates this insensitivity. We identified decreased transcriptional and functional responses to TGF-ß in CD8(+) DGK-ζ(-/-) T cells but preserved TGF-ß-mediated conversion of naïve DGK-ζ(-/-) CD4(+) T cells to a regulatory T cell phenotype. Decreased CD8(+) T cell responsiveness to TGF-ß did not result from impaired canonical TGF-ß signal transduction, because similar levels of TGF-ß-R and intracellular Smad components were identified in WT and DGK-ζ(-/-) CD8(+) T cells, and TGF-ß-mediated activation of Smad2 was unchanged. Instead, an enhanced TCR signal strength was responsible for TGF-ß insensitivity, because (i) loss of DGK-ζ conferred resistance to TGF-ß-mediated inhibition of Erk phosphorylation, (ii) TGF-ß insensitivity could be recapitulated by exogenous addition of the DAG analog PMA, and (iii) TGF-ß sensitivity could be observed in DGK-ζ-deficient T cells at limiting dilutions of TCR stimulation. These data indicate that enhanced TCR signal transduction in the absence of DGK-ζ makes T cells relatively insensitive to TGF-ß, in a manner independent of Smads, a finding with practical implications in the development of immunotherapies that target TGF-ß.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diacylglycerol Kinase/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Transforming Growth Factor beta/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Diacylglycerol Kinase/genetics , Mice , Mice, Knockout , Receptors, Antigen, T-Cell/genetics , Signal Transduction/genetics , Smad2 Protein/genetics , Smad2 Protein/immunology , Transforming Growth Factor beta/genetics
16.
Biochem Biophys Res Commun ; 463(3): 434-9, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26036574

ABSTRACT

Histamine and TGF-ß, major mediators secreted by mast cells, are involved in skin inflammation and play critical roles in the pathogenesis of systemic sclerosis. However, the roles of signaling mechanisms in the development of skin fibrosis remain largely unclear. Here we show that histamine suppressed the expression of α smooth muscle actin (αSMA), a marker of myofibroblasts, induced by TGF-ß1 in skin fibroblasts. Histamine H1-receptor (H1R), but not H2-receptor (H2R) or H4-receptor (H4R), was expressed on skin fibroblasts at both mRNA and protein levels. Interestingly, an H1R antagonist, but not H2R or H4R antagonists, antagonized the histamine-mediated suppression of αSMA expression by TGF-ß1. Correspondingly, phosphorylated Smad2 was detected after treatment with TGF-ß1, whereas the addition of histamine inhibited this phosphorylation. Taken together, histamine-H1R decreased TGF-ß1-mediated Smad2 phosphorylation and inhibited differentiation of skin fibroblasts into myofibroblasts.


Subject(s)
Fibroblasts/cytology , Histamine/immunology , Myofibroblasts/cytology , Receptors, Histamine H1/immunology , Skin/cytology , Transforming Growth Factor beta1/immunology , Actins/genetics , Cell Differentiation , Cell Line , Cell Line, Tumor , Fibroblasts/immunology , Gene Expression Regulation , Humans , Myofibroblasts/immunology , Phosphorylation , Receptors, Histamine H1/genetics , Skin/immunology , Smad2 Protein/chemistry , Smad2 Protein/immunology
17.
J Immunol ; 194(2): 709-18, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25505291

ABSTRACT

Macrophages are responsible for the control of inflammation and healing, and their malfunction results in cardiometabolic disorders. TGF-ß is a pleiotropic growth factor with dual (protective and detrimental) roles in atherogenesis. We have previously shown that in human macrophages, TGF-ß1 activates Smad2/3 signaling and induces a complex gene expression program. However, activated genes were not limited to known Smad2/3-dependent ones, which prompted us to study TGF-ß1-induced signaling in macrophages in detail. Analysis of Id3 regulatory sequences revealed a novel enhancer, located between +4517 and 4662 bp, but the luciferase reporter assay demonstrated that this enhancer is not Smad2/3 dependent. Because Id3 expression is regulated by Smad1/5 in endothelial cells, we analyzed activation of Smad1/5 in macrophages. We demonstrate here for the first time, to our knowledge, that TGF-ß1, but not BMPs, activates Smad1/5 in macrophages. We show that an ALK5/ALK1 heterodimer is responsible for the induction of Smad1/5 signaling by TGF-ß1 in mature human macrophages. Activation of Smad1/5 by TGF-ß1 induces not only Id3, but also HAMP and PLAUR, which contribute to atherosclerotic plaque vulnerability. We suggest that the balance between Smad1/5- and Smad2/3-dependent signaling defines the outcome of the effect of TGF-ß on atherosclerosis where Smad1/5 is responsible for proatherogenic effects, whereas Smad2/3 regulate atheroprotective effects of TGF-ß.


Subject(s)
Macrophages/immunology , Plaque, Atherosclerotic/immunology , Signal Transduction/immunology , Smad1 Protein/immunology , Smad5 Protein/immunology , Transforming Growth Factor beta1/immunology , Activin Receptors, Type II/immunology , Bone Morphogenetic Proteins/immunology , Cells, Cultured , Hepcidins/immunology , Humans , Inhibitor of Differentiation Proteins/immunology , Macrophages/pathology , Neoplasm Proteins/immunology , Plaque, Atherosclerotic/pathology , Protein Serine-Threonine Kinases/immunology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/immunology , Receptors, Urokinase Plasminogen Activator/immunology , Smad2 Protein/immunology , Smad3 Protein/immunology
18.
Immunity ; 41(3): 427-439, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25238097

ABSTRACT

Tumor-reactive T cells become unresponsive in advanced tumors. Here we have characterized a common mechanism of T cell unresponsiveness in cancer driven by the upregulation of the transcription factor Forkhead box protein P1 (Foxp1), which prevents CD8⁺ T cells from proliferating and upregulating Granzyme-B and interferon-γ in response to tumor antigens. Accordingly, Foxp1-deficient lymphocytes induced rejection of incurable tumors and promoted protection against tumor rechallenge. Mechanistically, Foxp1 interacted with the transcription factors Smad2 and Smad3 in preactivated CD8⁺ T cells in response to microenvironmental transforming growth factor-ß (TGF-ß), and was essential for its suppressive activity. Therefore, Smad2 and Smad3-mediated c-Myc repression requires Foxp1 expression in T cells. Furthermore, Foxp1 directly mediated TGF-ß-induced c-Jun transcriptional repression, which abrogated T cell activity. Our results unveil a fundamental mechanism of T cell unresponsiveness different from anergy or exhaustion, driven by TGF-ß signaling on tumor-associated lymphocytes undergoing Foxp1-dependent transcriptional regulation.


Subject(s)
Forkhead Transcription Factors/immunology , Neoplasms/immunology , Repressor Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Transforming Growth Factor beta/immunology , Tumor Escape/immunology , Adoptive Transfer , Animals , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Granzymes/biosynthesis , Interferon-gamma/biosynthesis , JNK Mitogen-Activated Protein Kinases/biosynthesis , JNK Mitogen-Activated Protein Kinases/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Signal Transduction/immunology , Smad2 Protein/immunology , Smad3 Protein/immunology , T-Lymphocytes, Cytotoxic/transplantation , Transcription, Genetic , Transcriptional Activation , Tumor Microenvironment/immunology
19.
Dev Comp Immunol ; 47(2): 197-204, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25092146

ABSTRACT

In the present study, we found that recombinant grass carp IL-1ß (rgcIL-1ß) simultaneously up-regulated grass carp IL-1ß (gcIL-1ß) and TGF-ß1 (gcTGF-ß1) expression via NF-κB and MAPK signaling in grass carp head kidney leukocytes (HKLs), promoting us to clarify whether TGF-ß1 is an effective antagonist in IL-1ß expression and activity. Our results showed that a stimulation of gcIL-1ß on its own expression was noted within 6 h, but gcTGF-ß1 neutralizing antibody prolonged gcIL-1ß autostimulation up to 12 h, indicating a possible inhibitory role of gcTGF-ß1 in regulating gcIL-1ß effect. This notion was reinforced by the fact that recombinant grass carp TGF-ß1 (rgcTGF-ß1) could impede rgcIL-1ß-induced gcIL-1ß gene expression and secretion in a reciprocal manner. Further studies revealed that rgcTGF-ß1 was able to attenuate rgcIL-1ß-induced mRNA expression of its own receptor signaling molecules and the activation of NF-κB. By contrast, rgcIL-1ß significantly amplified rgcTGF-ß1-mediated gcTGF-ß1 type I receptor (ALK5) expression and Smad2 phosphorylation in the same cell model. Taken together, these data shed light on an intrinsic mechanism for controlling inflammatory response by the reciprocal interaction between TGF-ß1 and IL-1ß in teleost.


Subject(s)
Fish Proteins/immunology , Head Kidney/immunology , Interleukin-1beta/immunology , Leukocytes/immunology , Protein Serine-Threonine Kinases/immunology , Receptors, Transforming Growth Factor beta/immunology , Signal Transduction/immunology , Transforming Growth Factor beta1/immunology , Animals , Antibodies, Neutralizing/pharmacology , Carps , Feedback, Physiological , Fish Proteins/genetics , Gene Expression Regulation , Head Kidney/cytology , Interleukin-1beta/genetics , Interleukin-1beta/pharmacology , Leukocytes/cytology , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/genetics , Smad2 Protein/genetics , Smad2 Protein/immunology , Transforming Growth Factor beta1/genetics
20.
J Immunol ; 192(10): 4560-4570, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24711618

ABSTRACT

Mesenchymal stem cells (MSCs) have been suggested to participate in immune regulation and airway repair/remodeling. TGF-ß1 is critical in the recruitment of stem/progenitor cells for tissue repair, remodeling, and cell differentiation. In this study, we sought to investigate the role of TGF-ß1 in MSC migration in allergic asthma. We examined nestin expression (a marker for MSCs) and TGF-ß1 signaling activation in airways in cockroach allergen extract (CRE)-induced mouse models. Compared with control mice, there were increased nestin(+) cells in airways and higher levels of active TGF-ß1 in serum and p-Smad2/3 expression in lungs of CRE-treated mice. Increased activation of TGF-ß1 signaling was also found in CRE-treated MSCs. We then assessed MSC migration induced by conditioned medium from CRE-challenged human epithelium in air/liquid interface culture in Transwell assays. MSC migration was stimulated by epithelial-conditioned medium, but was significantly inhibited by either TGF-ß1-neutralizing Ab or TßR1 inhibitor. Intriguingly, increased migration of MSCs from blood and bone marrow to the airway was also observed after systemic injection of GFP(+) MSCs and from bone marrow of Nes-GFP mice following CRE challenge. Furthermore, TGF-ß1-neutralizing Ab inhibited the CRE-induced MSC recruitment, but promoted airway inflammation. Finally, we investigated the role of MSCs in modulating CRE-induced T cell response and found that MSCs significantly inhibited CRE-induced inflammatory cytokine secretion (IL-4, IL-13, IL-17, and IFN-γ) by CD4(+) T cells. These results suggest that TGF-ß1 may be a key promigratory factor in recruiting MSCs to the airways in mouse models of asthma.


Subject(s)
Allergens/immunology , Asthma/immunology , Cell Movement/immunology , Cockroaches , Transforming Growth Factor beta1/immunology , Allergens/toxicity , Animals , Asthma/chemically induced , Asthma/pathology , Cell Movement/drug effects , Cytokines/immunology , Disease Models, Animal , Female , Humans , Male , Mice , Smad2 Protein/immunology , Smad3 Protein/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...