Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Fish Shellfish Immunol ; 113: 208-215, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33864946

ABSTRACT

The Smad protein family is an important medium for transducing BMP-Smads signals, and which have been proved that their important role in regulating shell biomineralization in Pinctada fucata martensii in our previous study. The members of TGF-ß superfamily were involved in innate immunity in vertebrates and invertebrates, and Smad regulatory networks construct a balanced immune system. However, little is known about the role of Smad1/5 in immunity in P. f. martensii. The present study shows that the tissue distribution and the expression profiles of Smad1/5 at developmental stages suggested its wide distribution and crucial role in development at embryonic stages other than larval stage; the increased expression of bone morphogenetic proteins 2 (BMP2), Smad4, Smad1/5 and MSX mRNAs at mantle tissue after LPS and Poly (I:C) challenged implied the potential immune role of Smad1/5 and BMP2-Smad signals to defense against bacterial and virus infections; the reduced expression of immune gene nuclear factor kappa-B (NF-κB), matrix metalloproteinase (MMP), interleukin 17 (IL-17), CuZn-superoxide dismutase (CuZn-SOD), tissue inhibitors of metalloproteinase (TIMP) and lipopolysaccharide-induced TNF-α factor (LITAF) mRNA following knockdown of Smad1/5 indicated that Smad1/5 can regulate their expression via BMP2-Smads pathway in the immunity process; the up-regulated expression of Smad1/5 and BMP2-Smad signals genes, and immune genes during wound healing indicated that Smad1/5 and BMP2-Smad signals genes may be involved in wound healing collaborated with immune genes via a different and complex Smads signaling pathway. These results indicated Smad1/5 could regulate innate immunity via BMP2-Smads signal pathway, and which provided new insights into the relationship between BMP2-Smads signal pathway and mantle immunity.


Subject(s)
Immunity, Innate/genetics , Pinctada/genetics , Pinctada/immunology , Signal Transduction/immunology , Smad Proteins/immunology , Animals , Gene Expression Profiling , Nacre/immunology , Smad Proteins/genetics
2.
J Environ Pathol Toxicol Oncol ; 40(2): 11-21, 2021.
Article in English | MEDLINE | ID: mdl-33822513

ABSTRACT

Global industrialization not only improved the quality life of millions but also paved the way to solving many health problems. One among them is allergic asthma, which affects approximately 20% of the global population. Poor air quality is the major culprit in allergic asthma, which not only affects the individual's health, but also impairs his or her life quality and that of family members. Asthma is a chronic pulmonary inflammatory disease characterized by excess mucus production, airway hyperresponsiveness, and bronchoconstriction. Inhalation of corticosteroids, leukotriene modifiers, and ß-adrenergic agonists is one treatment prescribed to control the symptoms of asthma, but there is still no effective cure. Phytochemicals such as carotenoids, phenolics, alkaloids, and nitrogen and organosulfur compounds are proven to possess immense pharmacological properties. Betalain is one such phytochemical present in plants of the order Caryophyllales. It is a water-soluble nitrogen-based pigment proven to possess antimicrobial, antioxidant, anti-inflammatory, hepatoprotective, antilipidemic, antidiabetic, and anticancer properties. We examined the curative potential of betalain against allergic asthma in a mouse model. Betalain treatment effectively decreased lung weight and infiltration of inflammatory cells in BAL fluid, and lowered IgE, eotaxin, and cytokine levels in asthma-induced mice. It also improved pulmonary mechanics and decreased oxidative stress and nitric oxide levels. Betalain significantly decreased gene expression of TGF-ß and its downstream signaling Smad proteins. Lung histology confirmed that betalain protected the lung tissue of mice from ovalbumin-induced allergic asthma. Overall, our results show that betalain is a potent antiallergic drug that effectively protects mice from ovalbumin-induced allergic asthma. With further research, it can be prescribed as a treatment for asthma in humans.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Asthma/drug therapy , Betalains/therapeutic use , Smad Proteins/immunology , Transforming Growth Factor beta1/immunology , Allergens , Animals , Anti-Asthmatic Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Asthma/immunology , Asthma/pathology , Asthma/physiopathology , Betalains/pharmacology , Cytokines/immunology , Disease Models, Animal , Female , Immunoglobulin E/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Lung/physiopathology , Mice, Inbred BALB C , Ovalbumin , Signal Transduction/drug effects , Smad Proteins/genetics , Transforming Growth Factor beta1/genetics
3.
Int Immunopharmacol ; 78: 106064, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31838448

ABSTRACT

Transforming growth factor (TGF)-ß/Smad signalling plays a central role in the pathogenesis of peritoneal fibrosis related to peritoneal dialysis (PD). Parthenolide (PTL), a naturally occurring phytochemical, is isolated from the shoots of feverfew (Tanacetum parthenium) and displays analgesia, anti-inflammation and anticancer activities. In this study, we examined the therapeutic potential of PTL on PD-related peritoneal fibrosis induced by daily intraperitoneal injection of 4.25% dextrose-containing PD fluid (PDF) in vivo and TGF-ß1-induced epithelial-mesenchymal transition (EMT) in vitro. PTL was administered daily before PDF injection or after 14 days of PDF injection. Both PTL treatments showed a protective effect on peritoneal fibrosis and prevented peritoneal dysfunction. Similarly, PTL suppressed the expression of fibrotic markers (fibronectin and collagen I) and restored the expression of the epithelial marker (E-cadherin) in TGF-ß1-treated HMrSV5 cells. Furthermore, PTL inhibited TGF-ß1-induced Smad2 and Smad3 phosphorylation and nuclear translocation but did not influence Smad1/5/9 phosphorylation or activate other downstream signalling pathways of TGF-ß1, including AKT, extracellular signal-regulated kinase (ERK) or p38. In conclusion, PTL treatment may represent an effective and novel therapy for PD-associated peritoneal fibrosis by suppressing the TGF-ß/Smad pathway.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/drug therapy , Sesquiterpenes/pharmacology , Signal Transduction/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cell Line , Dialysis Solutions/administration & dosage , Dialysis Solutions/adverse effects , Disease Models, Animal , Drug Evaluation, Preclinical , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/immunology , Female , Humans , Male , Mice , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/immunology , Peritoneal Fibrosis/pathology , Peritoneum/cytology , Peritoneum/drug effects , Peritoneum/immunology , Peritoneum/pathology , Phosphorylation/drug effects , Phosphorylation/immunology , Sesquiterpenes/therapeutic use , Signal Transduction/immunology , Smad Proteins/immunology , Smad Proteins/metabolism , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism
4.
Biochem Biophys Res Commun ; 522(3): 743-748, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31791581

ABSTRACT

Rheumatoid arthritis is a chronic and systemic autoimmune disease, which affects approximately 1% of the adult population worldwide. The present study investigated the therapeutic effect of theacrine (TC) on arthritis and its mechanisms in Freund's incomplete adjuvant (FIA)-induced SD rats. Rats were randomly divided into 5 groups: i) healthy control; ii) model; iii) positive control with methotrexate (MTX); iv) treatment with 12.5 mg/kg TC; and v) treatment with 25.0 mg/kg TC. The apparent scores, including changes in body weights, degree of paw swelling and arthritis indicators, were analyzed to evaluate the anti-chronic inflammatory effect of TC. The levels of interleukin (IL)-6 and transforming growth factor-ß (TGF-ß) in serum were measured by enzyme-linked immunosorbent assay. The protein and RNA expression levels of the critical factors in rats were measured to elucidate the mechanisms responsible for chronic inflammation and to verify molecular indexes of chronic inflammatory conditions. TC notably suppressed the severity of FIA-induced rat by attenuating the apparent scores, animal weight and inflammatory indexes in the 25 mg/kg TC group compared with the FIA rat model. Furthermore, TC significantly decreased the levels of IL-6 and increased the levels of TGF-ß. Histopathological examinations indicated that TC rescued the synovial hyperplasia and inflammatory cell infiltration in joint tissues. In addition, TC enhanced TGF-ß-mediated shifts in inflammatory marker expression in joint tissue. Overall, the present study demonstrated that TC exerted a superior anti-arthritic effect via the suppression of IL-6 and the activation of TGF-ß by the TGF-ß/SMAD pathway.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Smad Proteins/immunology , Transforming Growth Factor beta/immunology , Uric Acid/analogs & derivatives , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Chronic Disease , Freund's Adjuvant , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Joints/drug effects , Joints/immunology , Joints/pathology , Lipids , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Smad Proteins/analysis , Transforming Growth Factor beta/analysis , Uric Acid/therapeutic use
5.
Sci Signal ; 12(599)2019 09 17.
Article in English | MEDLINE | ID: mdl-31530731

ABSTRACT

Transforming growth factor-ß (TGF-ß) is produced by tumors, and increased amounts of this cytokine in the tumor microenvironment and serum are associated with poor patient survival. TGF-ß-mediated suppression of antitumor T cell responses contributes to tumor growth and survival. However, TGF-ß also has tumor-suppressive activity; thus, dissecting cell type-specific molecular effects may inform therapeutic strategies targeting this cytokine. Here, using human peripheral and tumor-associated lymphocytes, we investigated how tumor-derived TGF-ß suppresses a key antitumor function of CD4+ T cells, interferon-γ (IFN-γ) production. Suppression required the expression and phosphorylation of Smad proteins in the TGF-ß signaling pathway, but not their nuclear translocation, and depended on oxygen availability, suggesting a metabolic basis for these effects. Smad proteins were detected in the mitochondria of CD4+ T cells, where they were phosphorylated upon treatment with TGF-ß. Phosphorylated Smad proteins were also detected in the mitochondria of isolated tumor-associated lymphocytes. TGF-ß substantially impaired the ATP-coupled respiration of CD4+ T cells and specifically inhibited mitochondrial complex V (ATP synthase) activity. Last, inhibition of ATP synthase alone was sufficient to impair IFN-γ production by CD4+ T cells. These results, which have implications for human antitumor immunity, suggest that TGF-ß targets T cell metabolism directly, thus diminishing T cell function through metabolic paralysis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Mitochondria/immunology , Neoplasms/immunology , Oxygen Consumption/immunology , Transforming Growth Factor beta/immunology , Adenosine Triphosphate/immunology , Adenosine Triphosphate/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Humans , Interferon-gamma/metabolism , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/immunology , Mitochondrial Proton-Translocating ATPases/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation/immunology , Signal Transduction/immunology , Smad Proteins/immunology , Smad Proteins/metabolism , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/metabolism , Tumor Microenvironment/immunology
6.
Mol Med Rep ; 20(4): 3709-3718, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31485676

ABSTRACT

Chronic pancreatitis (CP) is characterized by recurrent pancreatic injury, resulting in inflammation and fibrosis. Currently, there are no drugs for the treatment of pancreatic fibrosis associated with CP. Piperine, a natural alkaloid found in black pepper, has been reported to show anti­inflammatory, anti­oxidative, and antitumor activities. Although piperine exhibits numerous properties in regards to the regulation of diverse diseases, the effects of piperine on CP have not been established. To investigate the effects of piperine on CP in vivo, we induced CP in mice through the repetitive administration of cerulein (50 µg/kg) six times at 1­h intervals, 5 times per week, for a total of 3 weeks. In the pre­treatment groups, piperine (1, 5, or 10 mg/kg) or corn oil were administrated orally at 1 h before the first cerulein injection, once a day, 5 times a week, for a total of 3 weeks. In the post­treatment groups, piperine (10 mg/kg) or corn oil was administered orally at 1 or 2 week after the first cerulein injection. Pancreases were collected for histological analysis. In addition, pancreatic stellate cells (PSCs) were isolated to examine the anti­fibrogenic effects and regulatory mechanisms of piperine. Piperine treatment significantly inhibited histological damage in the pancreas, increased the pancreatic acinar cell survival, reduced collagen deposition and reduced pro­inflammatory cytokines and chemokines. In addition, piperine treatment reduced the expression of fibrotic mediators, such as α­smooth muscle actin (α­SMA), collagen, and fibronectin 1 in the pancreas and PSCs. Moreover, piperine treatment reduced the production of transforming growth factor (TGF)­ß in the pancreas and PSCs. Furthermore, piperine treatment inhibited TGF­ß­induced pSMAD2/3 activation but not pSMAD1/5 in the PSCs. These findings suggest that piperine treatment ameliorates pancreatic fibrosis by inhibiting TGF­ß/SMAD2/3 signaling during CP.


Subject(s)
Alkaloids/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Benzodioxoles/therapeutic use , Pancreatitis, Chronic/drug therapy , Piperidines/therapeutic use , Polyunsaturated Alkamides/therapeutic use , Smad Proteins/immunology , Transforming Growth Factor beta/immunology , Animals , Disease Models, Animal , Female , Fibrosis , Mice , Mice, Inbred C57BL , Pancreas/drug effects , Pancreas/immunology , Pancreas/pathology , Pancreatitis, Chronic/immunology , Pancreatitis, Chronic/pathology , Signal Transduction/drug effects
7.
J Autoimmun ; 104: 102314, 2019 11.
Article in English | MEDLINE | ID: mdl-31416681

ABSTRACT

The TGF-ß superfamily of cytokines plays pivotal roles in the regulation of immune responses protecting against or contributing to diseases, such as, allergy, autoimmunity and cancer. Activin-A, a member of the TGF-ß superfamily, was initially identified as an inducer of follicle-stimulating hormone secretion. Extensive research over the past decades illuminated fundamental roles for activin-A in essential biologic processes, including embryonic development, stem cell maintenance and differentiation, haematopoiesis, cell proliferation and tissue fibrosis. Activin-A signals through two type I and two type II receptors which, upon ligand binding, activate their kinase activity, phosphorylate the SMAD2 and 3 intracellular signaling mediators that form a complex with SMAD4, translocate to the nucleus and activate or silence gene expression. Most immune cell types, including macrophages, dendritic cells (DCs), T and B lymphocytes and natural killer cells have the capacity to produce and respond to activin-A, although not in a similar manner. In innate immune cells, including macrophages, DCs and neutrophils, activin-A exerts a broad range of pro- or anti-inflammatory functions depending on the cell maturation and activation status and the spatiotemporal context. Activin-A also controls the differentiation and effector functions of Th cell subsets, including Th9 cells, TFH cells, Tr1 Treg cells and Foxp3+ Treg cells. Moreover, activin-A affects B cell responses, enhancing mucosal IgA secretion and inhibiting pathogenic autoantibody production. Interestingly, an array of preclinical and clinical studies has highlighted crucial functions of activin-A in the initiation, propagation and resolution of human diseases, including autoimmune diseases, such as, systemic lupus erythematosus, rheumatoid arthritis and pulmonary alveolar proteinosis, in allergic disorders, including allergic asthma and atopic dermatitis, in cancer and in microbial infections. Here, we provide an overview of the biology of activin-A and its signaling pathways, summarize recent studies pertinent to the role of activin-A in the modulation of inflammation and immunity, and discuss the potential of targeting activin-A as a novel therapeutic approach for the control of inflammatory diseases.


Subject(s)
Activins/immunology , Autoimmune Diseases/immunology , Hypersensitivity/immunology , Neoplasms/immunology , Active Transport, Cell Nucleus/immunology , Animals , Autoimmune Diseases/pathology , Autoimmune Diseases/therapy , Cell Nucleus/immunology , Cell Nucleus/pathology , Dendritic Cells , Humans , Hypersensitivity/pathology , Hypersensitivity/therapy , Leukocytes/immunology , Leukocytes/pathology , Neoplasm Proteins/immunology , Neoplasms/pathology , Neoplasms/therapy , Smad Proteins/immunology
8.
BMC Complement Altern Med ; 19(1): 14, 2019 Jan 10.
Article in English | MEDLINE | ID: mdl-30630477

ABSTRACT

BACKGROUND: Coreopsis tinctoria Nutt is an ethnomedicine widely used in Xinjiang, China. It is consumed as a herbal tea by local Uyghur people to treat high blood pressure and diarrhea. Our previous study confirmed that the ethyl acetate extract of Coreopsis tinctoria (AC) had a protective effect on diabetic nephropathy (DN) in an in vivo experiment. Here we aim to elucidate the protective mechanism of AC and marein, the main ingredient in Coreopsis tinctoria on renal fibrosis and inflammation in vitro under high glucose (HG) conditions. METHODS: A HG-induced barrier dysfunction model in rat mesangial cells (HBZY-1) was established. The cells were exposed to AC and marein and/or HG for 24 h. Then, the renal protective effects of AC and marein via transforming growth factor-ß1 (TGF-ß1)/Smads, AMP-activated kinase protein (AMPK), and nuclear factor kappa beta (NF-κB) signaling were assessed. RESULTS: Both AC and marein suppressed rat mesangial cell hyperplasia and significantly attenuated the expression of HG-disrupted fibrotic and inflammatory proteins in HBZY-1 cells. It was also confirmed that AC and marein remarkably attenuated HG-induced renal inflammation and fibrosis by regulating the AMPK, TGF-ß1/Smads, and NF-κB signaling pathways. CONCLUSION: These results indicated that AC and marein may delay the progression of DN, at least in part, by suppressing HG-induced renal inflammation and fibrosis. Marein may be one of the bioactive compounds in AC.


Subject(s)
Coreopsis/chemistry , Drugs, Chinese Herbal/pharmacology , Glucose/adverse effects , Kidney Diseases/immunology , NF-kappa B/immunology , Protein Kinases/immunology , Smad Proteins/immunology , Transforming Growth Factor beta1/immunology , AMP-Activated Protein Kinase Kinases , Animals , Chalcones/pharmacology , Fibrosis/drug therapy , Fibrosis/genetics , Fibrosis/immunology , Humans , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Mesangial Cells/drug effects , Mesangial Cells/immunology , NF-kappa B/genetics , Protein Kinases/genetics , Rats , Signal Transduction/drug effects , Smad Proteins/genetics , Transforming Growth Factor beta1/genetics
9.
Int Rev Immunol ; 37(4): 192-205, 2018.
Article in English | MEDLINE | ID: mdl-29672174

ABSTRACT

T lymphocytes represent an important part of adaptive immune system undertaking different functions to regulate immune responses. CD4+ T cells are the most important activator cells in inflammatory conditions. Depending on the type of induced cells and inflamed sites, expression and activity of different subtypes of helper T cells are changed. Recent studies have confirmed the existence of a new subset of helper T lymphocytes called Th9. Naive T cells can differentiate into Th9 subtypes if they are exposed simultaneously by interleukin (IL) 4 and transforming growth factor ß and also secondary activation of a complicated network of transcription factors such as interferon regulatory factor 4 (IRF4) and Smads which are essential for adequate induction of this phenotype. Th9 cells specifically produce interleukin 9 and their probable roles in promoting intestinal inflammation are being investigated in human subjects and experimental models of ulcerative colitis (UC). Recently, infiltration of Th9 cells, overexpression of IL-9, and certain genes associated with Th9 differentiation have been demonstrated in inflammatory microenvironment of UC. Intestinal oversecretion of IL-9 protein is likely to break down epithelial barriers and compromise tolerance to certain commensal microorganisms which leads to inflammation. Th9 pathogenicity has not yet been adequately explored in UC and they are far from being considered as inflammatory cells in this milieu, therefore precise understanding the role of these newly identified cells in particular their potential role in gut pathogenesis may enable us to develop novel therapeutic approaches for inflammatory bowel disease. So, this article tries to discuss the latest knowledge on the above-mentioned field.


Subject(s)
Colitis, Ulcerative/immunology , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Cell Differentiation/immunology , Colitis, Ulcerative/pathology , Colon/immunology , Colon/metabolism , Colon/microbiology , Colon/pathology , Humans , Immune Tolerance/immunology , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Interleukin-9/immunology , Interleukin-9/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Signal Transduction/immunology , Smad Proteins/immunology , Smad Proteins/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
10.
J Infect ; 76(4): 406-416, 2018 04.
Article in English | MEDLINE | ID: mdl-29391143

ABSTRACT

OBJECTIVES: Cystic echinococcosis (CE) is a zoonotic disease caused by Echinococcus granulosus (Eg) infection. Th9 cells are reported to be involved in the immune responses in CE patients. This study aims to investigate the role of TGF-ß/Smad pathway in the regulation of Th9 cells in CE patients. METHODS: Using Western blot analysis, flow cytometry, qPCR, immunohistochemistry, ELISA and MTT assay, we measured the expression levels of TGF-ß/Smad, PU.1, IRF-4, and IL-9 in CE patients. RESULTS: The levels of TGF-ß, p-Samd3, PU.1 and IL-9 were elevated in the liver of CE patients. IL-9 and IL-9R expressions were also elevated in the infected liver tissue, and IL-9 level was positively correlated with the liver inflammation. The levels of IL-9, IL-4, TGF-ß and IL-10 in the supernatant were also significantly increased after stimulating hepatic lymphocytes of CE patients with Eg antigen B. After blocking the TGF-ß pathway signaling in vitro, PU.1 and IL-9 were obviously reduced. CONCLUSIONS: IL-9 may aggravate the inflammatory response in the liver of CE patients. The TGF-ß/Smad signaling pathway is activated, and the signaling pathway may promote the differentiation of Th9 cells and IL-9 expression in active CE patients.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Echinococcosis/immunology , Interleukin-9/immunology , Signal Transduction , Smad Proteins/immunology , Transforming Growth Factor beta/immunology , Adult , Animals , Antigens, Helminth/pharmacology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Cytokines/immunology , Echinococcus granulosus , Hepatocytes/drug effects , Hepatocytes/immunology , Humans , Liver/immunology , Liver/parasitology , Male , Middle Aged , Proto-Oncogene Proteins/immunology , Trans-Activators/immunology , Up-Regulation
11.
Nat Immunol ; 18(7): 800-812, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28504697

ABSTRACT

An imbalance in the lineages of immunosuppressive regulatory T cells (Treg cells) and the inflammatory TH17 subset of helper T cells leads to the development of autoimmune and/or inflammatory disease. Here we found that TAZ, a coactivator of TEAD transcription factors of Hippo signaling, was expressed under TH17 cell-inducing conditions and was required for TH17 differentiation and TH17 cell-mediated inflammatory diseases. TAZ was a critical co-activator of the TH17-defining transcription factor RORγt. In addition, TAZ attenuated Treg cell development by decreasing acetylation of the Treg cell master regulator Foxp3 mediated by the histone acetyltransferase Tip60, which targeted Foxp3 for proteasomal degradation. In contrast, under Treg cell-skewing conditions, TEAD1 expression and sequestration of TAZ from the transcription factors RORγt and Foxp3 promoted Treg cell differentiation. Furthermore, deficiency in TAZ or overexpression of TEAD1 induced Treg cell differentiation, whereas expression of a transgene encoding TAZ or activation of TAZ directed TH17 cell differentiation. Our results demonstrate a pivotal role for TAZ in regulating the differentiation of Treg cells and TH17 cells.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Cell Differentiation/immunology , Colitis/immunology , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Intracellular Signaling Peptides and Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Animals , Arthritis, Rheumatoid/immunology , Case-Control Studies , Chromatin Immunoprecipitation , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Flow Cytometry , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , HEK293 Cells , HeLa Cells , Histone Acetyltransferases/metabolism , Humans , Immunoblotting , Lysine Acetyltransferase 5 , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proteasome Endopeptidase Complex/metabolism , Real-Time Polymerase Chain Reaction , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Sjogren's Syndrome/immunology , Smad Proteins/immunology , Smad Proteins/metabolism , TEA Domain Transcription Factors , Trans-Activators/metabolism , Transcription Factors/immunology , Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins
12.
Fish Shellfish Immunol ; 67: 129-140, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28546027

ABSTRACT

The proteins of Smad family are critical components of the TGF-ß superfamily signal pathway. In this paper, we cloned two intracellular mediators of TGF-ß signaling, Smad3 and Smad5, from the pearl mussel Hyriopsis cumingii. The full length cDNA of HcSmad3 and HcSmad5 were 2052 bp and 1908 bp and encoded two polypeptides of 418 and 461amino acid residues, respectively. The deduced amino acid of HcSmad3 and HcSmad5 possessed two putative conserved domains, MH1 and MH2, a conserved phosphorylation motif SSXS at the carboxyl-terminal. The two Smad genes were detected muscle, mantle, hepatopancreas and gill, but with a very low level in heamocytes. The transcripts of Smad3 and Smad5 were up-regulated in hemocytes and hepatopancreas after A. hydrophila and PGN stimulation. However, the expression of Smad3 and Smad5 were only up-regulated in hepatopancreas after A. hydrophila stimulation. The transcripts of Smad3 and Smad5 had a slight change in hepatopancreas after PGN stimulation. The transcripts of HcSmad3 showed very little increase and HcSmad5 mRNA significantly up-regulated after wounding.


Subject(s)
Immunity, Innate/genetics , Smad Proteins/genetics , Smad Proteins/immunology , Unionidae/genetics , Unionidae/immunology , Wound Healing/immunology , Aeromonas hydrophila/physiology , Amino Acid Sequence , Animals , Base Sequence , Phylogeny , Sequence Alignment , Smad Proteins/chemistry
13.
Sci Signal ; 9(418): ra27, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26956486

ABSTRACT

Transforming growth factor-ß (TGF-ß) is an immunosuppressive cytokine that inhibits the proinflammatory functions of T cells, and it is a major factor in abrogating T cell activity against tumors. Canonical TGF-ß signaling results in the activation of Smad proteins, which are transcription factors that regulate target gene expression. We found that the cell surface molecule platelet endothelial cell adhesion molecule-1 (PECAM-1) facilitated noncanonical (Smad-independent) TGF-ß signaling in T cells. Subcutaneously injected tumor cells that are dependent on TGF-ß-mediated suppression of immunity for growth grew more slowly in PECAM-1(-/-) mice than in their wild-type counterparts. T cells isolated from PECAM-1(-/-) mice demonstrated relative insensitivity to the TGF-ß-dependent inhibition of interferon-γ (IFN-γ) production, granzyme B synthesis, and cellular proliferation. Similarly, human T cells lacking PECAM-1 demonstrated decreased sensitivity to TGF-ß in a manner that was partially restored by reexpression of PECAM-1. Co-incubation of T cells with TGF-ß and a T cell-activating antibody resulted in PECAM-1 phosphorylation on an immunoreceptor tyrosine-based inhibitory motif (ITIM) and the recruitment of the inhibitory Src homology 2 (SH2) domain-containing tyrosine phosphatase-2 (SHP-2). Such conditions also induced the colocalization of PECAM-1 with the TGF-ß receptor complex as identified by coimmunoprecipitation, confocal microscopy, and proximity ligation assays. These studies indicate a role for PECAM-1 in enhancing the inhibitory functions of TGF-ß in T cells and suggest that therapeutic targeting of the PECAM-1-TGF-ß inhibitory axis represents a means to overcome TGF-ß-dependent immunosuppression within the tumor microenvironment.


Subject(s)
Platelet Endothelial Cell Adhesion Molecule-1/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta/immunology , Amino Acid Motifs , Animals , Granzymes/genetics , Granzymes/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Smad Proteins/genetics , Smad Proteins/immunology , Transforming Growth Factor beta/genetics
14.
BMC Pulm Med ; 15: 13, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25888222

ABSTRACT

BACKGROUND: Interleukin-27 (IL-27) is a multifunctional cytokine with both pro-inflammatory and immunoregulatory functions. At present, the role of IL-27 in pulmonary fibrosis remains unknown. METHODS: In this study, we observed the expression of IL-27/IL-27R in a mouse model of bleomycin (BLM)-induced pulmonary fibrosis. We verified the role of IL-27 using hematoxylin and eosin as well as Masson's staining methods and measuring the content of hydroxyproline as well as collagen I and III. We assessed the differentiation of T lymphocytes in the spleen and measured the concentration of cytokines in bronchoalveolar lavage fluid (BALF) and the expression level of relevant proteins in the JAK/STAT and TGF-ß/Smad signaling pathways in lung tissue. RESULTS: Increased IL-27 expression in BLM-induced pulmonary fibrosis was noted. IL-27 treatment may alleviate pulmonary fibrosis and increase the survival of mice. IL-27 inhibited the development of CD4(+) IL-17(+), CD4(+) IL-4(+) T, and CD4(+) Foxp3(+) cells and the secretion of IL-17, IL-4, IL-6, and TGF-ß. IL-27 induced the production of CD4(+) IL-10(+) and CD4(+) INF-γ(+) T cells. IL-27 decreased the levels of phosphorylated STAT1, STAT3, STAT5, Smad1, and Smad3 but increased the level of SOCS3. CONCLUSIONS: This study demonstrates that IL-27 potentially attenuates BLM-induced pulmonary fibrosis by regulating Th17 differentiation and cytokine secretion.


Subject(s)
Cell Differentiation/immunology , Cytokines/immunology , Interleukins/genetics , Lung/immunology , Pulmonary Fibrosis/immunology , Receptors, Cytokine/genetics , Th17 Cells/immunology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Blotting, Western , Bronchoalveolar Lavage Fluid/immunology , Cell Differentiation/drug effects , Cytokines/drug effects , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interleukins/immunology , Interleukins/pharmacology , Janus Kinases/drug effects , Janus Kinases/immunology , Lung/drug effects , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Real-Time Polymerase Chain Reaction , Receptors, Cytokine/immunology , Receptors, Interleukin , Reverse Transcriptase Polymerase Chain Reaction , STAT Transcription Factors/drug effects , STAT Transcription Factors/immunology , Signal Transduction , Smad Proteins/drug effects , Smad Proteins/immunology , Spleen/cytology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Suppressor of Cytokine Signaling Proteins/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Th17 Cells/drug effects , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/immunology
15.
Trends Immunol ; 36(3): 150-60, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25725583

ABSTRACT

Whereas type I interferons (IFNs) have critical roles in protection from pathogens, excessive IFN responses contribute to pathology in both acute and chronic settings, pointing to the importance of balancing activating signals with regulatory mechanisms that appropriately tune the response. Here we review evidence for an integrated network of negative regulators of IFN production and action, which function at all levels of the activating and effector signalling pathways. We propose that the aim of this extensive network is to limit tissue damage while enabling an IFN response that is temporally appropriate and of sufficient magnitude. Understanding the architecture and dynamics of this network, and how it differs in distinct tissues, will provide new insights into IFN biology and aid the design of more effective therapeutics.


Subject(s)
Immunity, Innate , Interferon Type I/immunology , Intracellular Signaling Peptides and Proteins/immunology , MicroRNAs/immunology , Smad Proteins/immunology , Ubiquitin-Protein Ligases/immunology , Dendritic Cells/immunology , Feedback, Physiological , Gene Expression Regulation/immunology , Gene Regulatory Networks/immunology , Humans , Interferon Type I/genetics , Interleukins/genetics , Interleukins/immunology , Intracellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , Receptors, Cytokine/genetics , Receptors, Cytokine/immunology , Signal Transduction , Smad Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Viruses/immunology
16.
Eur J Immunol ; 44(11): 3283-94, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25142202

ABSTRACT

Bone morphogenetic protein 4 (BMP4) is a multifunctional growth factor that belongs to the TGF-ß superfamily. The role of BMP4 in lung diseases is not fully understood. Here, we demonstrate that BMP4 was upregulated in lungs undergoing lipopolysaccharide (LPS)-induced inflammation, and in airway epithelial cells treated with LPS or TNF-α. BMP4 mutant (BMP4(+/-) ) mice presented with more severe lung inflammation in response to LPS or Pseudomonas aeruginosa, and lower bacterial load compared with that in BMP4(+/+) mice. Knockdown of BMP4 by siRNA increased LPS and TNF-α-induced IL-8 expression in 16HBE human airway epithelial cells and in primary human bronchial epithelial cells. Similarly, peritoneal macrophages from BMP4(+/-) mice produced greater levels of TNF-α and keratinocyte chemoattractant (KC) upon LPS treatment compared with cells from BMP4(+/+) mice. Administration of exogenous BMP4 attenuated the upregulation of TNF-α, IL-8, or KC induced by LPS and/or TNF-α in airway epithelial cells, and peritoneal macrophages. Finally, partial deficiency of BMP4 in BMP4(+/-) mice protected the animals from restrictive lung function reduction upon chronic LPS exposure. These results indicate that BMP4 plays an important anti-inflammatory role, controlling the strength and facilitating the resolution of acute lung inflammation; yet, BMP4 also contributes to lung function impairment during chronic lung inflammation.


Subject(s)
Bone Morphogenetic Protein 4/immunology , Lung/immunology , Pneumonia, Bacterial/immunology , Pseudomonas Infections/immunology , Animals , Bone Morphogenetic Protein 4/genetics , Cells, Cultured , Epithelial Cells/immunology , Humans , Inflammation/chemically induced , Inflammation/immunology , Inflammation Mediators , Interleukin-8/biosynthesis , Lipopolysaccharides/pharmacology , Lung/microbiology , Lung Injury/prevention & control , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pneumonia, Bacterial/microbiology , Pseudomonas aeruginosa/immunology , RNA Interference , RNA, Small Interfering , Signal Transduction/immunology , Smad Proteins/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
17.
Int Immunopharmacol ; 20(1): 248-57, 2014 May.
Article in English | MEDLINE | ID: mdl-24631515

ABSTRACT

Alveolar echinococcosis (AE) is a severe parasitic disease caused by the infection of Echinococcus multilocularis (Em). Very little is known on the relationship between TGF-ß/Smad signaling pathway and Treg/Th17 balance in the infected liver at different periods after Em infection. Using qRT-PCR, immunohistochemistry, flow cytometry and CBA assay, we measured the expression levels of TGF-ß, Smad2/3/7, ROR-γt, Foxp3, IL-17, IL-10 and percentages of Th17 cells and Treg cells in mouse AE model, from day 2 to day 270 after infection. In the early stage of infection (day 2 to day 30), Smad7 was up-regulated and the TGF-ß pathway was inactivated. In the middle stage of infection (day 30 to day 90), TGF-ß and Smad2/3 were up-regulated. And levels of Treg cells, Foxp3, Th17 cells, RORγt, IL-17, IL-10 and IL-6 were significantly increased. In the late stage of infection (day 90 to day 270), Treg cells, Foxp3, TGF-ß and IL-10 maintained at high levels whereas Th17 cells and IL-17 decreased significantly. TGF-ß/Smad signaling pathway was activated during the chronic infection. Our data suggest that there were Treg/Th17 imbalance in the middle and especially in the late stage of Em infection and that Treg/Th17 imbalance may be regulated by TGF-ß/Smad signaling pathway. Treg and Th17 subsets may be involved in regulating immune tolerance and tissue inflammation, and facilitating the long-term survival of Em in the host.


Subject(s)
Echinococcosis/immunology , Smad Proteins/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta/immunology , Animals , Echinococcosis/pathology , Echinococcus multilocularis , Female , Forkhead Transcription Factors/genetics , Interleukin-10/immunology , Interleukin-17/immunology , Liver/drug effects , Liver/immunology , Liver/pathology , Mice, Inbred BALB C , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , RNA, Messenger/metabolism , Receptors, Transforming Growth Factor beta/immunology , Signal Transduction , Smad Proteins/genetics , Transforming Growth Factor beta/genetics
18.
Protein Pept Lett ; 21(1): 45-51, 2014.
Article in English | MEDLINE | ID: mdl-23919379

ABSTRACT

Myostatin plays negative roles in muscle development. To block the inhibitory effects of myostatin on myogenesis, a 759 bp single chain variable fragment antibody (scFv) against myostatin was constructed and expressed in Escherichia coli. ELISA detection showed that the scFv could bind to myostatin, and change of the scFv N-terminal peptides decreased its binding affinity. MTT assay and cell morphology demonstrated that the cell number and viability of the C2C12 myoblast were enhanced by the scFv. Meanwhile, the scFv significantly inhibited the myostatin-induced expression of cyclin-dependent kinase inhibitor p21 and Smad binding element-luciferase activity. H2O2 increased the expression of Muscle RING Finger 1 (MuRF1) and Muscle Atrophy F-box (MAFbx) in myoblasts as well as myostatin and MuRF1 in myotubes, and the scFv significantly decreased the H2O2-elevated expression of these genes. Conclusively, the scFv we developed could antagonize the inhibitory effects of myostatin on myogenesis through Smad pathway and regulation of p21, MuRF1 and MAFbx gene expression. The scFv may have application in the therapy of muscular dystrophy and improvement of animal meat production.


Subject(s)
Muscle Development/physiology , Myoblasts/immunology , Myostatin/immunology , Single-Chain Antibodies/immunology , Antibody Affinity/immunology , Base Sequence , Cell Survival , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Humans , Hydrogen Peroxide/pharmacology , Muscle Proteins/biosynthesis , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscular Dystrophies/therapy , Myoblasts/cytology , Protein Binding/physiology , SKP Cullin F-Box Protein Ligases/biosynthesis , Single-Chain Antibodies/biosynthesis , Smad Proteins/biosynthesis , Smad Proteins/immunology , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/biosynthesis
19.
Immunology ; 139(1): 1-10, 2013 May.
Article in English | MEDLINE | ID: mdl-23347175

ABSTRACT

A balanced immune response requires combating infectious assaults while striving to maintain quiescence towards the self. One of the central players in this process is the pleiotropic cytokine transforming growth factor-ß (TGF-ß), whose deficiency results in spontaneous systemic autoimmunity in mice. The dominant function of TGF-ß is to regulate the peripheral immune homeostasis, particularly in the microbe-rich and antigen-rich environment of the gut. To maintain intestinal integrity, the epithelial cells, myeloid cells and lymphocytes that inhabit the gut secrete TGF-ß, which acts in both paracrine and autocrine fashions to activate its signal transducers, the SMAD transcription factors. The SMAD pathway regulates the production of IgA by B cells, maintains the protective mucosal barrier and promotes the balanced differentiation of CD4(+) T cells into inflammatory T helper type 17 cells and suppressive FOXP3(+) T regulatory cells. While encounters with pathogenic microbes activate SMAD proteins to evoke a protective inflammatory immune response, SMAD activation and synergism with immunoregulatory factors such as the vitamin A metabolite retinoic acid enforce immunosuppression toward commensal microbes and innocuous food antigens. Such complementary context-dependent functions of TGF-ß are achieved by the co-operation of SMAD proteins with distinct dominant transcription activators and accessory chromatin modifiers. This review highlights recent advances in unravelling the molecular basis for the multi-faceted functions of TGF-ß in the gut that are dictacted by fluid orchestrations of SMADs and their myriad partners.


Subject(s)
B-Lymphocytes/immunology , Immunity, Mucosal/physiology , Intestinal Mucosa/immunology , Smad Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Autocrine Communication/immunology , B-Lymphocytes/cytology , Humans , Immune Tolerance/physiology , Immunoglobulin A/immunology , Intestinal Mucosa/cytology , Mice , Paracrine Communication/immunology , T-Lymphocytes, Regulatory/cytology , Th17 Cells/cytology , Transforming Growth Factor beta/immunology
20.
Eur J Immunol ; 42(7): 1796-803, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22585622

ABSTRACT

Notch proteins play an important role in embryonic development and cell-fate decisions. Notch influences also the activation and differentiation of peripheral T cells. Here, we investigated whether Notch signaling modulates the response of effector T cells to regulatory T (Treg) cells. Pre-exposure of CD4(+) CD25(-) effector T cells to the Notch ligands Delta-4 and Jagged-1, but not Delta-1, increases significantly effector T-cell sensitivity to Treg cell-mediated suppression through upregulation of TGF-ßRII expression and increased levels of the phosphorylated form of the Smad 3 protein. This effect is relieved by anti-TGF-ß Abs. We demonstrate that HES (hairy and enhancer of split), the main transcription factor downstream of Notch, induces strong transactivation of TGF-ßRII by binding the TGF-ßRII promoter through its DNA-binding domain. Thus, the crosstalk between Notch and the TGF-ß pathway leads to potentiation of the suppressive effect of Treg cells.


Subject(s)
Protein Serine-Threonine Kinases/immunology , Receptor, Notch1/immunology , Receptors, Transforming Growth Factor beta/immunology , Smad Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Basic Helix-Loop-Helix Transcription Factors/immunology , Calcium-Binding Proteins/immunology , Humans , Immunoblotting , Intercellular Signaling Peptides and Proteins/immunology , Jagged-1 Protein , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Membrane Proteins/immunology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA/chemistry , RNA/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Reverse Transcriptase Polymerase Chain Reaction , Serrate-Jagged Proteins , Signal Transduction , Smad Proteins/genetics , T-Lymphocytes, Regulatory/cytology , Transcriptional Activation , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...