Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 700
Filter
1.
Fish Shellfish Immunol ; 149: 109564, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38631439

ABSTRACT

Grass carp reovirus (GCRV) infections and hemorrhagic disease (GCHD) outbreaks are typically seasonally periodic and temperature-dependent, yet the molecular mechanism remains unclear. Herein, we depicted that temperature-dependent IL-6/STAT3 axis was exploited by GCRV to facilitate viral replication via suppressing type Ⅰ IFN signaling. Combined multi-omics analysis and qPCR identified IL-6, STAT3, and IRF3 as potential effector molecules mediating GCRV infection. Deploying GCRV challenge at 18 °C and 28 °C as models of resistant and permissive infections and switched to the corresponding temperatures as temperature stress models, we illustrated that IL-6 and STAT3 expression, genome level of GCRV, and phosphorylation of STAT3 were temperature dependent and regulated by temperature stress. Further research revealed that activating IL-6/STAT3 axis enhanced GCRV replication and suppressed the expression of IFNs, whereas blocking the axis impaired viral replication. Mechanistically, grass carp STAT3 inhibited IRF3 nuclear translocation via interacting with it, thus down-regulating IFNs expression, restraining transcriptional activation of the IFN promoter, and facilitating GCRV replication. Overall, our work sheds light on an immune evasion mechanism whereby GCRV facilitates viral replication by hijacking IL-6/STAT3 axis to down-regulate IFNs expression, thus providing a valuable reference for targeted prevention and therapy of GCRV.


Subject(s)
Carps , Fish Diseases , Interferon Type I , Interleukin-6 , Reoviridae Infections , Reoviridae , STAT3 Transcription Factor , Signal Transduction , Virus Replication , Animals , Fish Diseases/immunology , Fish Diseases/virology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/metabolism , Reoviridae Infections/immunology , Reoviridae Infections/veterinary , Reoviridae/physiology , Carps/immunology , Carps/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Interferon Type I/immunology , Interferon Type I/genetics , Fish Proteins/genetics , Fish Proteins/immunology , Immunity, Innate/genetics
2.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167151, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38565387

ABSTRACT

Immunoglobulin A vasculitis (IgAV) is the most common vasculitis of childhood. Disordered immune responses play important roles in its pathogenesis, but the comprehensive immune profile of the disease and the underlying mechanisms are still largely unknown. Here we found a potential disease biomarker cold inducible RNA binding protein (CIRP) in our pediatric IgAV cohort. Serum CIRP level in these patients were elevated and positively correlated with the increased early memory (CD45RA+CD62L+CD95+) T cells revealed using multicolor flow cytometry. Immune phenotyping of the patients showed they had more activated T cells with higher IL6Ra expression. T cell culture experiment showed CIRP further activated both human CD4+ and CD8+ T cells as indicated by increased perforin secretion and phosphorylation of STAT3. Blockade of IL6Rα attenuated CIRP-induced T cell toxicity in vitro. RNA-sequencing data further supported CIRP stimulation promoted human T cell activation and migration, fueled inflammation through the JAK-STAT signaling pathway. Therefore, IL6Ra-mediated T cell activation by extracellular CIRP may contribute to pathogenesis of IgAV in children, both CIRP and IL6Ra could be new therapeutic targets for IgAV.


Subject(s)
Lymphocyte Activation , RNA-Binding Proteins , Receptors, Interleukin-6 , STAT3 Transcription Factor , Humans , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/immunology , Receptors, Interleukin-6/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/immunology , Child , Male , Female , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/immunology , Lymphocyte Activation/immunology , Signal Transduction , IgA Vasculitis/immunology , IgA Vasculitis/pathology , IgA Vasculitis/metabolism , Adolescent , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Hepatitis A Virus Cellular Receptor 2
3.
Mol Immunol ; 170: 46-56, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615627

ABSTRACT

Peritoneal B cells can be divided into B1 cells (CD11b+CD19+) and B2 cells (CD11b-CD19+) based on CD11b expression. B1 cells play a crucial role in the innate immune response by producing natural antibodies and cytokines. B2 cells share similar traits with B1 cells, influenced by the peritoneal environment. However, the response of both B1 and B2 cells to the same stimuli in the peritoneum remains uncertain. We isolated peritoneal B1 and B2 cells from mice and assessed differences in Interleukin-10(IL-10) secretion, apoptosis, and surface molecule expression following exposure to LPS and Interleukin-21(IL-21). Our findings indicate that B1 cells are potent IL-10 producers, possessing surface molecules with an IgMhiCD43+CD21low profile, and exhibit a propensity for apoptosis in vitro. Conversely, B2 cells exhibit lower IL-10 production and surface markers characterized as IgMlowCD43-CD21hi, indicative of some resistance to apoptosis. LPS stimulates MAPK phosphorylation in B1 and B2 cells, causing IL-10 production. Furthermore, LPS inhibits peritoneal B2 cell apoptosis by enhancing Bcl-xL expression. Conversely, IL-21 has no impact on IL-10 production in these cells. Nevertheless, impeding STAT3 phosphorylation permits IL-21 to increase IL-10 production in peritoneal B cells. Moreover, IL-21 significantly raises apoptosis levels in these cells, a process independent of STAT3 phosphorylation and possibly linked to reduced Bcl-xL expression. This study elucidates the distinct functional and response profiles of B1 and B2 cells in the peritoneum to stimuli like LPS and IL-21, highlighting their differential roles in immunological responses and B cell diversity.


Subject(s)
Apoptosis , Interleukin-10 , Interleukins , Lipopolysaccharides , Peritoneum , Interleukins/immunology , Interleukins/metabolism , Animals , Lipopolysaccharides/pharmacology , Lipopolysaccharides/immunology , Mice , Interleukin-10/immunology , Interleukin-10/metabolism , Apoptosis/drug effects , Apoptosis/immunology , Peritoneum/immunology , Peritoneum/cytology , B-Lymphocyte Subsets/immunology , Mice, Inbred C57BL , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/immunology , B-Lymphocytes/immunology , CD11b Antigen/metabolism , CD11b Antigen/immunology , bcl-X Protein/metabolism , bcl-X Protein/immunology , Phosphorylation/drug effects , Antigens, CD19/immunology , Antigens, CD19/metabolism
4.
Cell Rep ; 42(7): 112806, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37440406

ABSTRACT

This study identifies interleukin-6 (IL-6)-independent phosphorylation of STAT3 Y705 at the early stage of infection with several viruses, including influenza A virus (IAV). Such activation of STAT3 is dependent on the retinoic acid-induced gene I/mitochondrial antiviral-signaling protein/spleen tyrosine kinase (RIG-I/MAVS/Syk) axis and critical for antiviral immunity. We generate STAT3Y705F/+ knockin mice that display a remarkably suppressed antiviral response to IAV infection, as evidenced by impaired expression of several antiviral genes, severe lung tissue injury, and poor survival compared with wild-type animals. Mechanistically, STAT3 Y705 phosphorylation restrains IAV pathogenesis by repressing excessive production of interferons (IFNs). Blocking phosphorylation significantly augments the expression of type I and III IFNs, potentiating the virulence of IAV in mice. Importantly, knockout of IFNAR1 or IFNLR1 in STAT3Y705F/+ mice protects the animals from lung injury and reduces viral load. The results indicate that activation of STAT3 by Y705 phosphorylation is vital for establishment of effective antiviral immunity by suppressing excessive IFN signaling induced by viral infection.


Subject(s)
Influenza A virus , Orthomyxoviridae Infections , STAT3 Transcription Factor , Animals , Mice , Antiviral Agents , Immunity, Innate , Interferons , Receptors, Interferon , Signal Transduction , Orthomyxoviridae Infections/immunology , STAT3 Transcription Factor/immunology
5.
Clin Exp Immunol ; 212(2): 107-116, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36652220

ABSTRACT

The STAT3 story has almost 30 years of evolving history. First identified in 1994 as a pro-inflammatory transcription factor, Signal Transducer and Activator of Transcription 3 (STAT3) has continued to be revealed as a quintessential pleiotropic signalling module spanning fields including infectious diseases, autoimmunity, vaccine responses, metabolism, and malignancy. In 2007, germline heterozygous dominant-negative loss-of-function variants in STAT3 were discovered as the most common cause for a triad of eczematoid dermatitis with recurrent skin and pulmonary infections, first described in 1966. This finding established that STAT3 plays a critical non-redundant role in immunity against some pathogens, as well as in the connective tissue, dental and musculoskeletal systems. Several years later, in 2014, heterozygous activating gain of function germline STAT3 variants were found to be causal for cases of early-onset multiorgan autoimmunity, thereby underpinning the notion that STAT3 function needed to be regulated to maintain immune homeostasis. As we and others continue to interrogate biochemical and cellular perturbations due to inborn errors in STAT3, we will review our current understanding of STAT3 function, mechanisms of disease pathogenesis, and future directions in this dynamic field.


Subject(s)
Immunity , STAT3 Transcription Factor , Humans , Autoimmunity/genetics , Autoimmunity/immunology , Mutation/genetics , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Immunity/genetics , Immunity/immunology , Immune System Diseases/genetics , Immune System Diseases/immunology
6.
Int Immunopharmacol ; 113(Pt A): 109424, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36461589

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease of unknown cause and characterized by excessive proliferation of fibroblasts and the irregular remodeling of extracellular matrix (ECM), which ultimately cause the severe distortion of the alveolar architecture. The median survival of IPF patients is 2-5 years. IPF patients are predominantly infiltrated by M2 macrophages during the course of disease development and progression. Predominantly accumulation of M2 macrophages accelerates fibrosis progression by secreting multiple cytokines that promote fibroblast to myofibroblast transition. In the process of M2 macrophage polarization, JAK2/STAT3 signaling plays a key role, thus, targeting activated macrophages to inhibit the pro-fibrotic phenotype is considered as an approach to the potential treatment of IPF. Tacrolimus is a macrolide antibiotic that as a specific inhibitor of T-lymphocyte function and has been used widely as an immunosuppressant in human organ transplantation. In this study we explored the potential effect and mechanism of tacrolimus on pulmonary fibrosis in vivo and vitro. Here, we found that tacrolimus is capable of suppressing M2 macrophages polarization by inhibiting pro-fibrotic factors secreted by M2 macrophages. This effect further alleviates M2-induced myofibroblast activation, thus resulting in a decline of collagen deposition, pro-fibrotic cytokines secretion, recovering of lung function, ultimately relieving the progression of fibrosis in vivo. Mechanistically, we found that tacrolimus can inhibit the activation of JAK2/STAT3 signaling by targeting JAK2. Our findings indicate a potential anti-fibrotic effect of tacrolimus by regulating macrophage polarization and might be meaningful in clinical settings.


Subject(s)
Bleomycin , Idiopathic Pulmonary Fibrosis , Macrophages , Tacrolimus , Humans , Bleomycin/adverse effects , Cytokines , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/immunology , Janus Kinase 2/immunology , Macrophages/drug effects , Macrophages/immunology , STAT3 Transcription Factor/immunology , Tacrolimus/pharmacology , Tacrolimus/therapeutic use
7.
Cell Mol Gastroenterol Hepatol ; 14(4): 789-811, 2022.
Article in English | MEDLINE | ID: mdl-35809803

ABSTRACT

BACKGROUND & AIMS: MUC1 is abnormally expressed in colorectal cancer, including colitis-associated colorectal cancer (CAC), but its role in tumorigenesis is unclear. This study investigated MUC1's effects in murine models of colitis and CAC and elucidated mechanisms of action. METHODS: Colitis and CAC were induced in mice by exposure to dextran sodium sulfate or azoxymethane plus dextran sodium sulphate. Clinical parameters, immune cell infiltration, and tumor development were monitored throughout disease progression. Experiments in knockout mice and bone marrow chimeras were combined with an exploration of immune cell abundance and function. RESULTS: Deficiency of Muc1 suppressed inflammation, inhibited tumor progression, increased abundance of CD8+ T lymphocytes, and reduced abundance of macrophages in colon tumors. Bone marrow chimeras showed promotion of CAC was primarily mediated by Muc1-expressing hematopoietic cells, and that MUC1 promoted a pro-tumoral immunosuppressive macrophage phenotype within tumors. Mechanistic studies revealed that Muc1 deficiency remarkably reduced interleukin-6 levels in the colonic tissues and tumors that was mainly produced by infiltrating macrophages at day 21, 42, and 85. In bone marrow-derived macrophages, MUC1 promoted responsiveness to chemoattractant and promoted activation into a phenotype with high Il6 and Ido1 expression, secreting factors which inhibited CD8+ T cell proliferation. MUC1 potently drives macrophages to produce interleukin-6, which in turn drives a pro-tumorigenic activation of signal transducer and activator of transcription 3 in colon epithelial tumor and stromal cells, ultimately increasing the occurrence and development of CAC. CONCLUSIONS: Our findings provide cellular and molecular mechanisms for the pro-tumorigenic functions of MUC1 in the inflamed colon. Therapeutic strategies to inhibit MUC1 signal transduction warrant consideration for the prevention or therapy of CAC.


Subject(s)
Colitis-Associated Neoplasms , Interleukin-6 , Macrophage Activation , Mucin-1 , STAT3 Transcription Factor , Animals , Azoxymethane/toxicity , Carcinogenesis , Chemotactic Factors , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colitis-Associated Neoplasms/genetics , Colitis-Associated Neoplasms/immunology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Dextran Sulfate/toxicity , Interleukin-6/genetics , Interleukin-6/immunology , Macrophage Activation/genetics , Macrophage Activation/immunology , Mice , Mice, Knockout , Mucin-1/genetics , Mucin-1/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology
8.
Int J Hematol ; 115(6): 816-825, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35275353

ABSTRACT

Acquired chronic pure red cell aplasia (PRCA) develops idiopathically or in association with other medical conditions, including T cell large granular lymphocytic leukemia (T-LGLL) and thymoma. T cell dysregulation is considered a cardinal pathogenesis of PRCA, but genetic-phenotypic associations in T cell abnormalities are largely unclear. We evaluated an extended cohort of 90 patients with acquired PRCA, including 26 with idiopathic, 36 with T-LGLL-associated and 15 with thymoma-associated PRCA, for their T cell immuno-phenotypes, clonalities and STAT3 mutations. TCR repertoire skewing of CD8+ T cells was detected in 37.5% of idiopathic, 66.7% of T-LGLL-associated and 25% of thymoma-associated PRCA patients, and restriction to Vß1 was most prominent (41%). Clonalities of TCRß or γ chain and STAT3 mutational status were statistically associated (P = 0.0398), and they were detected in all three subtypes. The overall response rate to cyclosporin A was 73.9%, without significant difference by subtypes nor STAT3 mutational status. The T cell dysregulations, such as TCR repertoire skewing with predominant Vß1 usage, clonality and STAT3 mutations, were frequently found across the subtypes, and the close associations between them suggest that these T cell derangements reflect a common pathophysiological mechanism among these PRCA subtypes.


Subject(s)
Leukemia, Large Granular Lymphocytic , Red-Cell Aplasia, Pure , STAT3 Transcription Factor , Thymoma , Thymus Neoplasms , CD8-Positive T-Lymphocytes/pathology , Humans , Leukemia, Large Granular Lymphocytic/genetics , Leukemia, Large Granular Lymphocytic/immunology , Leukemia, Large Granular Lymphocytic/pathology , Mutation , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Red-Cell Aplasia, Pure/genetics , Red-Cell Aplasia, Pure/immunology , Red-Cell Aplasia, Pure/pathology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Thymoma/genetics , Thymoma/immunology , Thymus Neoplasms/immunology
9.
Biomed Environ Sci ; 35(2): 95-106, 2022 Feb 20.
Article in English | MEDLINE | ID: mdl-35197174

ABSTRACT

OBJECTIVE: This study aimed to investigate the effects of caprylic acid (C8:0) on lipid metabolism and inflammation, and examine the mechanisms underlying these effects in mice and cells. METHODS: Fifty-six 6-week-old male C57BL/6J mice were randomly allocated to four groups fed a high-fat diet (HFD) without or with 2% C8:0, palmitic acid (C16:0) or eicosapentaenoic acid (EPA). RAW246.7 cells were randomly divided into five groups: normal, lipopolysaccharide (LPS), LPS+C8:0, LPS+EPA and LPS+cAMP. The serum lipid profiles, inflammatory biomolecules, and ABCA1 and JAK2/STAT3 mRNA and protein expression were measured. RESULTS: C8:0 decreased TC and LDL-C, and increased the HDL-C/LDL-C ratio after injection of LPS. Without LPS, it decreased TC in mice ( P < 0.05). Moreover, C8:0 decreased the inflammatory response after LPS treatment in both mice and cells ( P < 0.05). Mechanistic investigations in C57BL/6J mouse aortas after injection of LPS indicated that C8:0 resulted in higher ABCA1 and JAK2/STAT3 expression than that with HFD, C16:0 and EPA, and resulted in lower TNF-α, NF-κB mRNA expression than that with HFD ( P < 0.05). In RAW 264.7 cells, C8:0 resulted in lower expression of pNF-κBP65 than that in the LPS group, and higher protein expression of ABCA1, p-JAK2 and p-STAT3 than that in the LPS and LPS+cAMP groups ( P < 0.05). CONCLUSION: Our studies demonstrated that C8:0 may play an important role in lipid metabolism and the inflammatory response, and the mechanism may be associated with ABCA1 and the p-JAK2/p-STAT3 signaling pathway.


Subject(s)
ATP Binding Cassette Transporter 1/immunology , Caprylates/administration & dosage , Inflammation/drug therapy , Janus Kinase 2/immunology , Lipid Metabolism/drug effects , Macrophages/drug effects , STAT3 Transcription Factor/immunology , ATP Binding Cassette Transporter 1/genetics , Animals , Caprylates/chemistry , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Humans , Inflammation/etiology , Inflammation/immunology , Inflammation/metabolism , Janus Kinase 2/genetics , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , STAT3 Transcription Factor/genetics , Signal Transduction
10.
Cell Rep ; 38(3): 110244, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35045292

ABSTRACT

The active form of vitamin D, 1,25-dihydroxyvitamin D3, induces a stable tolerogenic phenotype in dendritic cells (DCs). This process involves the vitamin D receptor (VDR), which translocates to the nucleus, binds its cognate genomic sites, and promotes epigenetic and transcriptional remodeling. In this study, we report the occurrence of vitamin D-specific DNA demethylation and transcriptional activation at VDR binding sites associated with the acquisition of tolerogenesis in vitro. Differentiation to tolerogenic DCs associates with activation of the IL-6-JAK-STAT3 pathway. We show that JAK2-mediated STAT3 phosphorylation is specific to vitamin D stimulation. VDR and the phosphorylated form of STAT3 interact with each other to form a complex with methylcytosine dioxygenase TET2. Most importantly, pharmacological inhibition of JAK2 reverts vitamin D-induced tolerogenic properties of DCs. This interplay among VDR, STAT3, and TET2 opens up possibilities for modulating DC immunogenic properties in clinics.


Subject(s)
DNA-Binding Proteins/immunology , Dendritic Cells/immunology , Dioxygenases/immunology , Immune Tolerance/immunology , Receptors, Calcitriol/immunology , STAT3 Transcription Factor/immunology , Cells, Cultured , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Dioxygenases/metabolism , Humans , Receptors, Calcitriol/metabolism , STAT3 Transcription Factor/metabolism
11.
Cancer Sci ; 113(2): 432-445, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34927311

ABSTRACT

Thrombospondin-1 (TSP1) is generally assumed to suppress the growth of osteosarcoma through inhibiting angiogenesis; however, it is unclear whether TSP1 could affect the antitumor immunity against osteosarcoma. We aimed to explore the immune-related tumor-promoting effects of TSP1 and decipher its underlying mechanism. First, we identified that TSP1 regulated programmed death-ligand 1 (PD-L1) expression, which was related to the CD8+ T cells anergy in osteosarcoma cells. The exact role of PD-L1 in the immunosuppressive effect of TSP1 was then further confirmed by the addition of the PD-L1 neutralizing Ab. With the addition of PD-L1 neutralizing Abs during cocultivation, the inhibition of CD8+ T cells was abolished to a certain extent. Further mechanistic investigations showed that TSP1-induced PD-L1 upregulation was achieved by activation of the signal transducer and activator of transcription 3 (STAT3) pathway. In vivo experiments also indicated that TSP1 overexpression could promote the growth of primary lesions, whereas TSP1 knockdown effectively inhibits the growth of the primary lesion as well as lung metastasis by restoring the antitumor immunity. Thrombospondin-1 knockdown combined with PD-L1 neutralizing Ab achieved a more pronounced antitumor effect. Taken together, our study showed that TSP1 upregulates PD-L1 by activating the STAT3 pathway and, therefore, impairs the antitumor immunity against osteosarcoma.


Subject(s)
B7-H1 Antigen/immunology , Bone Neoplasms/immunology , Immune Tolerance , Osteosarcoma/immunology , STAT3 Transcription Factor/immunology , Thrombospondin 1/immunology , Animals , Apoptosis , B7-H1 Antigen/genetics , Bone Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Mice , Osteosarcoma/pathology , Signal Transduction , Thrombospondin 1/genetics
12.
Article in English | WPRIM (Western Pacific) | ID: wpr-927639

ABSTRACT

OBJECTIVE@#This study aimed to investigate the effects of caprylic acid (C8:0) on lipid metabolism and inflammation, and examine the mechanisms underlying these effects in mice and cells.@*METHODS@#Fifty-six 6-week-old male C57BL/6J mice were randomly allocated to four groups fed a high-fat diet (HFD) without or with 2% C8:0, palmitic acid (C16:0) or eicosapentaenoic acid (EPA). RAW246.7 cells were randomly divided into five groups: normal, lipopolysaccharide (LPS), LPS+C8:0, LPS+EPA and LPS+cAMP. The serum lipid profiles, inflammatory biomolecules, and ABCA1 and JAK2/STAT3 mRNA and protein expression were measured.@*RESULTS@#C8:0 decreased TC and LDL-C, and increased the HDL-C/LDL-C ratio after injection of LPS. Without LPS, it decreased TC in mice ( P < 0.05). Moreover, C8:0 decreased the inflammatory response after LPS treatment in both mice and cells ( P < 0.05). Mechanistic investigations in C57BL/6J mouse aortas after injection of LPS indicated that C8:0 resulted in higher ABCA1 and JAK2/STAT3 expression than that with HFD, C16:0 and EPA, and resulted in lower TNF-α, NF-κB mRNA expression than that with HFD ( P < 0.05). In RAW 264.7 cells, C8:0 resulted in lower expression of pNF-κBP65 than that in the LPS group, and higher protein expression of ABCA1, p-JAK2 and p-STAT3 than that in the LPS and LPS+cAMP groups ( P < 0.05).@*CONCLUSION@#Our studies demonstrated that C8:0 may play an important role in lipid metabolism and the inflammatory response, and the mechanism may be associated with ABCA1 and the p-JAK2/p-STAT3 signaling pathway.


Subject(s)
Animals , Humans , Male , Mice , ATP Binding Cassette Transporter 1/immunology , Caprylates/chemistry , Cholesterol/metabolism , Diet, High-Fat/adverse effects , Inflammation/metabolism , Janus Kinase 2/immunology , Lipid Metabolism/drug effects , Macrophages/immunology , Mice, Inbred C57BL , STAT3 Transcription Factor/immunology , Signal Transduction
13.
Front Immunol ; 12: 767939, 2021.
Article in English | MEDLINE | ID: mdl-34858425

ABSTRACT

The tumor microenvironment (TME) is composed of a heterogenous population of cells that exist alongside the extracellular matrix and soluble components. These components can shape an environment that is conducive to tumor growth and metastatic spread. It is well-established that stromal cancer-associated fibroblasts (CAFs) in the TME play a pivotal role in creating and maintaining a growth-permissive environment for tumor cells. A growing body of work has uncovered that tumor cells recruit and educate CAFs to remodel the TME, however, the mechanisms by which this occurs remain incompletely understood. Recent studies suggest that the signal transducer and activator of transcription 3 (STAT3) is a key transcription factor that regulates the function of CAFs, and their crosstalk with tumor and immune cells within the TME. CAF-intrinsic STAT3 activity within the TME correlates with tumor progression, immune suppression and eventually the establishment of metastases. In this review, we will focus on the roles of STAT3 in regulating CAF function and their crosstalk with other cells constituting the TME and discuss the utility of targeting STAT3 within the TME for therapeutic benefit.


Subject(s)
Cancer-Associated Fibroblasts/immunology , Neoplasms/immunology , STAT3 Transcription Factor/immunology , Signal Transduction/immunology , Tumor Microenvironment/immunology , Cancer-Associated Fibroblasts/metabolism , Cell Communication/immunology , Disease Progression , Humans , Janus Kinases/immunology , Janus Kinases/metabolism , Neoplasms/metabolism , Neoplasms/therapy , Phosphorylation/immunology , STAT3 Transcription Factor/metabolism
14.
Front Immunol ; 12: 773013, 2021.
Article in English | MEDLINE | ID: mdl-34925343

ABSTRACT

Gardnerella vaginalis is associated with bacterial vaginosis (BV). The virulence factors produced by G. vaginalis are known to stimulate vaginal mucosal immune response, which is largely driven by activated macrophages. While Tilapia piscidin 4 (TP4), an antimicrobial peptide isolated from Nile tilapia, is known to display a broad range of antibacterial functions, it is unclear whether TP4 can affect macrophage polarization in the context of BV. In this study, we used the culture supernatants from G. vaginalis to stimulate differentiation of THP-1 and RAW264.7 cells to an M1 phenotype. The treatment activated the NF-κB/STAT1 signaling pathway, induced reactive nitrogen and oxygen species, and upregulated inflammatory mediators. We then treated the induced M1 macrophages directly with a non-toxic dose of TP4 or co-cultured the M1 macrophages with TP4-treated vaginal epithelial VK2 cells. The results showed that TP4 could not only decrease pro-inflammatory mediators in the M1 macrophages, but it also enriched markers of M2 macrophages. Further, we found that direct treatment with TP4 switched M1 macrophages toward a resolving M2c phenotype via the MAPK/ERK pathway and IL-10-STAT3 signaling. Conversely, tissue repair M2a macrophages were induced by TP4-treated VK2 cells; TP4 upregulated TSG-6 in VK2 cells, which subsequently activated STAT6 and M2a-related gene expression in the macrophages. In conclusion, our results imply that TP4 may be able to attenuate the virulence of G. vaginalis by inducing resolving M2c and tissue repair M2a macrophage polarizations, suggesting a novel strategy for BV therapy.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Gardnerella vaginalis , Macrophages/immunology , Vaginosis, Bacterial/immunology , Animals , Cell Line , Cichlids , Cytokines/immunology , Female , Humans , MAP Kinase Signaling System , Mice , Models, Biological , STAT3 Transcription Factor/immunology
15.
Front Immunol ; 12: 739219, 2021.
Article in English | MEDLINE | ID: mdl-34912331

ABSTRACT

Active form of vitamin D (VitD) enhances human innate immunity against Mycobacterium tuberculosis (Mtb) infection. Our previous studies showed that MIR337-3p was highly expressed in lymphocytes of tuberculosis (TB) patients. Here, we identified the mechanism of MIR337-3p in the regulation of fast-acting anti-TB immunity by inhibiting VitD-dependent antimicrobial response pathways. While high-level MIR337-3p expression was induced by mycobacterial infection in cellular models and mice, TB patients exhibited significantly increased MIR337-3p in CD14+ monocytes/macrophages, innate-like Vγ2+ T cells, and CD8+ lymphocytes containing natural killer (NK)/innate lymphoid cells. MIR337-3p promoted the mycobacterial entry/infection and replication/growth in host target cells: macrophages and lung epithelial cells. Such MIR337-3p-enhanced pathogenicity coincided with the MIR337-3p depression of VitD-dependent antimicrobial response of cytochrome P450, family 27, subfamily b, polypeptide 1 (CYP27B1)/Beta-defensin 4 (DEFB4A)/ cathelicidin antimicrobial peptide CAMP pathways. Surprisingly, single MIR337-3p species could specifically target both the Toll-like receptor 4 (TLR4) and signal transducer and activator of transcription 3 (STAT3) 3'-untranslated regions (UTRs) to depress the TLR4/MYD88 and STAT3 signals and impair either of the two signals inhibiting the VitD-dependent antimicrobial pathways in macrophages. Concurrently, human peripheral blood mononuclear cells (PBMCs) expressing high-level MIR337-3p exhibited a reduced ability of innate cell populations to mount fast-acting cellular immunity against intracellular mycobacterial infection. Furthermore, a higher expression of Mir337-3p after mycobacterial infection of mice coincided with much greater colony-forming unit (CFU) counts in lungs and even the death of infected animals, whereas Mir337-3p inhibitor treatment of infected mice reduced Mir337-3p levels and reversed Mir337-3p-mediated increases in CFU counts. Thus, TB-driven single MIR337-3p species could specifically target/impair both TLR4/MYD88 and STAT3 activation signals, inhibiting VitD-dependent antimicrobial response and fast-acting anti-TB immunity, leading to enhanced pathogenicity.


Subject(s)
Immunity, Innate/immunology , MicroRNAs/immunology , Mycobacterium tuberculosis/pathogenicity , Receptors, Calcitriol/immunology , Tuberculosis/immunology , Animals , Humans , Immunity, Innate/genetics , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
16.
Biochemistry (Mosc) ; 86(11): 1489-1501, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34906042

ABSTRACT

Bronchial asthma is a heterogeneous chronic inflammatory disease of airways. The studies of molecular and cellular mechanisms of bronchial asthma have established that a wide range of immune (T and B cells, eosinophils, neutrophils, macrophages, etc.) and structural (epithelial and endothelial) cells are involved in its pathogenesis. These cells are activated in response to external stimuli (bacteria, viruses, allergens, and other pollutants) and produce pro-inflammatory factors (cytokines, chemokines, metalloproteinases, etc.), which ultimately leads to the initiation of pathological processes in the lungs. Genes encoding transcription factors of the STAT family (signal transducer and activator of transcription), that includes seven representatives, are involved in the cell activation. Recent studies have shown that the transcription factor STAT3 plays an important role in the activation of the abovementioned cells, thus contributing to the development of asthma. In animal studies, selective inhibition of STAT3 significantly reduces the severity of lung inflammation, which indicates its potential as a therapeutic target. In this review, we describe the mechanisms of STAT3 activation and its role in polarization of Th2/Th17 cells and M2 macrophages, as well as in the dysfunction of endothelial cells, which ultimately leads to development of bronchial asthma symptoms, such as infiltration of neutrophils and eosinophils into the lungs, bronchial hyperreactivity, and the respiratory tract remodeling.


Subject(s)
Asthma/immunology , Leukocytes/immunology , Lung/immunology , STAT3 Transcription Factor/immunology , Animals , Asthma/pathology , Humans , Leukocytes/pathology , Lung/pathology
17.
Front Immunol ; 12: 663177, 2021.
Article in English | MEDLINE | ID: mdl-34867936

ABSTRACT

Dominant-negative mutations associated with signal transducer and activator of transcription 3 (STAT3) signaling, which controls epithelial proliferation in various tissues, lead to atopic dermatitis in hyper IgE syndrome. This dermatitis is thought to be attributed to defects in STAT3 signaling in type 17 helper T cell specification. However, the role of STAT3 signaling in skin epithelial cells remains unclear. We found that STAT3 signaling in keratinocytes is required to maintain skin homeostasis by negatively controlling the expression of hair follicle-specific keratin genes. These expression patterns correlated with the onset of dermatitis, which was observed in specific pathogen-free conditions but not in germ-free conditions, suggesting the involvement of Toll-like receptor-mediated inflammatory responses. Thus, our study suggests that STAT3-dependent gene expression in keratinocytes plays a critical role in maintaining the homeostasis of skin, which is constantly exposed to microorganisms.


Subject(s)
Hair Follicle/physiology , STAT3 Transcription Factor/physiology , Animals , Dermatitis, Atopic/etiology , Dermatitis, Atopic/genetics , Dermatitis, Atopic/immunology , Disease Models, Animal , Female , Gene Expression Regulation , Hair Follicle/immunology , Homeostasis , Humans , Keratinocytes/immunology , Keratinocytes/physiology , Keratins/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , Signal Transduction , Skin/immunology , Skin/microbiology , Skin Physiological Phenomena , Th17 Cells/immunology
18.
Front Immunol ; 12: 724609, 2021.
Article in English | MEDLINE | ID: mdl-34603297

ABSTRACT

STAT3 activates transcription of genes that regulate cell growth, differentiation, and survival of mammalian cells. Genetic deletion of Stat3 in T cells has been shown to abrogate Th17 differentiation, suggesting that STAT3 is a potential therapeutic target for Th17-mediated diseases. However, a major impediment to therapeutic targeting of intracellular proteins such as STAT3 is the lack of efficient methods for delivering STAT3 inhibitors into cells. In this study, we developed a novel antibody (SBT-100) comprised of the variable (V) region of a STAT3-specific heavy chain molecule and demonstrate that this 15 kDa STAT3-specific nanobody enters human and mouse cells, and induced suppression of STAT3 activation and lymphocyte proliferation in a concentration-dependent manner. To investigate whether SBT-100 would be effective in suppressing inflammation in vivo, we induced experimental autoimmune uveitis (EAU) in C57BL/6J mice by active immunization with peptide from the ocular autoantigen, interphotoreceptor retinoid binding protein (IRBP651-670). Analysis of the retina by fundoscopy, histological examination, or optical coherence tomography showed that treatment of the mice with SBT-100 suppressed uveitis by inhibiting expansion of pathogenic Th17 cells that mediate EAU. Electroretinographic (ERG) recordings of dark and light adapted a- and b-waves showed that SBT-100 treatment rescued mice from developing significant visual impairment observed in untreated EAU mice. Adoptive transfer of activated IRBP-specific T cells from untreated EAU mice induced EAU, while EAU was significantly attenuated in mice that received IRBP-specific T cells from SBT-100 treated mice. Taken together, these results demonstrate efficacy of SBT-100 in mice and suggests its therapeutic potential for human autoimmune diseases.


Subject(s)
Autoimmune Diseases/prevention & control , STAT3 Transcription Factor/immunology , Th17 Cells/immunology , Uveitis/prevention & control , Adoptive Transfer , Animals , Autoantigens/immunology , Autoantigens/metabolism , Autoimmune Diseases/immunology , Cell Proliferation/drug effects , Cytokines/metabolism , Disease Models, Animal , Electroretinography , Eye Proteins/immunology , Eye Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Retinol-Binding Proteins/immunology , Retinol-Binding Proteins/metabolism , STAT3 Transcription Factor/metabolism , Th17 Cells/pathology , Uveitis/immunology
19.
Front Immunol ; 12: 661323, 2021.
Article in English | MEDLINE | ID: mdl-34531850

ABSTRACT

Tumors evade the immune system by inducing inflammation. In melanoma, tumor-derived IL-1ß drives inflammation and the expansion of highly immunosuppressive myeloid-derived suppressor cells (MDSCs). Similar in many tumors, melanoma is also linked to the downstream IL-6/STAT3 axis. In this study, we observed that both recombinant and tumor-derived IL-1ß specifically induce pSTAT3(Y705), creating a tumor-autoinflammatory loop, which amplifies IL-6 signaling in the human melanoma cell line 1205Lu. To disrupt IL-1ß/IL-6/STAT3 axis, we suppressed IL-1ß-mediated inflammation by inhibiting the NOD-like receptor protein 3 (NLRP3) using OLT1177, a safe-in-humans specific NLRP3 oral inhibitor. In vivo, using B16F10 melanoma, OLT1177 effectively reduced tumor progression (p< 0.01); in primary tumors, OLT1177 decreased pSTAT3(Y705) by 82% (p<0.01) and II6 expression by 53% (p<0.05). Disruption of tumor-derived NLRP3, either pharmacologically or genetically, reduced STAT3 signaling in bone marrow cells. In PMN-MDSCs isolated from tumor-bearing mice treated with OLT1177, we observed significant reductions in immunosuppressive genes such as Pdcd1l1, Arg1, Il10 and Tgfb1. In conclusion, the data presented here show that the inhibition of NLRP3 reduces IL-1ß induction of pSTAT3(Y705) preventing expression of immunosuppressive genes as well as activity in PMN-MDSCs.


Subject(s)
Interleukin-1beta/immunology , Interleukin-6/immunology , Melanoma/immunology , Myeloid-Derived Suppressor Cells/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , STAT3 Transcription Factor/immunology , Animals , Cell Line, Tumor , Humans , Immune Tolerance/drug effects , Immune Tolerance/immunology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Melanoma/metabolism , Melanoma/pathology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Myeloid-Derived Suppressor Cells/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nitriles/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Burden/drug effects , Tumor Burden/immunology
20.
Int J Mol Sci ; 22(18)2021 Sep 11.
Article in English | MEDLINE | ID: mdl-34576006

ABSTRACT

Tumor immune escape is a common process in the tumorigenesis of non-small cell lung cancer (NSCLC) cells where programmed death ligand-1 (PD-L1) expression, playing a vital role in immunosuppression activity. Additionally, epidermal growth factor receptor (EGFR) phosphorylation activates Janus kinase-2 (JAK2) and signal transduction, thus activating transcription 3 (STAT3) to results in the regulation of PD-L1 expression. Chemotherapy with commercially available drugs against NSCLC has struggled in the prospect of adverse effects. Nobiletin is a natural flavonoid isolated from the citrus peel that exhibits anti-cancer activity. Here, we demonstrated the role of nobiletin in evasion of immunosuppression in NSCLC cells by Western blotting and real-time polymerase chain reaction methods for molecular signaling analysis supported by gene silencing and specific inhibitors. From the results, we found that nobiletin inhibited PD-L1 expression through EGFR/JAK2/STAT3 signaling. We also demonstrated that nobiletin exhibited p53-independent PD-L1 suppression, and that miR-197 regulates the expression of STAT3 and PD-L1, thereby enhancing anti-tumor immunity. Further, we evaluated the combination ability of nobiletin with an anti-PD-1 monoclonal antibody in NSCLC co-culture with peripheral blood mononuclear cells. Similarly, we found that nobiletin assisted the induction of PD-1/PD-L1 blockade, which is a key factor for the immune escape mechanism. Altogether, we propose nobiletin as a modulator of tumor microenvironment for cancer immunotherapy.


Subject(s)
B7-H1 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Flavones/pharmacology , Lung Neoplasms/immunology , MicroRNAs/immunology , Neoplasm Proteins/immunology , RNA, Neoplasm/immunology , STAT3 Transcription Factor/immunology , Signal Transduction/drug effects , Tumor Escape/drug effects , A549 Cells , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...