Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters











Publication year range
1.
Mol Immunol ; 171: 22-35, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38749236

ABSTRACT

OBJECTIVES: Systemic lupus erythematosus (SLE) is a multi-organ autoimmune disease, of which the pathogens is remains obscure. Viral infection, particularly Epstein Barr viru (EBV) infection, has been considered a common pathogenic factor. This study suggests that c-Maf may be an important target in T cell differentiation during SLE progression, providing a potentially new perspective on the role of viral infection in the pathogenesis of autoimmune diseases. METHODS: Cytokines of EBV-infected SLE patients were measured by ELISA and assessed in conjunction with their clinical data. IFN-α, c-Maf, and the differentiation of Th17/Treg cells in SLE patients and MRL/LPR mice were analyzed using FCM, WB, RT-PCR, etc. Following the infection of cells and mice with EBV or viral mimic poly (dA:dT), the changes of the aforementioned indicators were investigated. The relationship among IFN-α, STAT3, c-Maf and Th17 cells was determined by si-RNA technique. RESULTS: Many SLE patients are found to be complicated by viral infections; Further, studies have demonstrated that viral infection, especially EBV, is involved in SLE development. This study showed that viral infections might promote IFN-α secretion, inhibit c-Maf expression by activating STAT3, increase Th17 cell differentiation, and lead to the immune imbalance of Th17/Treg cells, thus playing a role in the onset and progression of SLE. CONCLUSION: This study demonstrates that EBV infections may contribute to SLE development by activating STAT3 through IFN-α, inhibiting c-Maf, and causing Th17/Treg immune imbalance. Our work provided a new insight into the pathogenesis and treatment of SLE.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , Interferon-alpha , Lupus Erythematosus, Systemic , Mice, Inbred MRL lpr , Proto-Oncogene Proteins c-maf , T-Lymphocytes, Regulatory , Th17 Cells , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/virology , Th17 Cells/immunology , Humans , Animals , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Infections/complications , T-Lymphocytes, Regulatory/immunology , Mice , Interferon-alpha/immunology , Interferon-alpha/metabolism , Female , Adult , Herpesvirus 4, Human/immunology , Proto-Oncogene Proteins c-maf/immunology , Proto-Oncogene Proteins c-maf/genetics , Male , Cell Differentiation/immunology , Disease Progression , Middle Aged , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/immunology , Young Adult
2.
PLoS Pathog ; 17(7): e1009732, 2021 07.
Article in English | MEDLINE | ID: mdl-34280251

ABSTRACT

We have recently demonstrated that the function of T follicular helper (Tfh) cells from lymph nodes (LN) of HIV-infected individuals is impaired. We found that these cells were unable to provide proper help to germinal center (GC)-B cells, as observed by altered and inefficient anti-HIV antibody response and premature death of memory B cells. The underlying molecular mechanisms of this dysfunction remain poorly defined. Herein, we have used a unique transcriptional approach to identify these molecular defects. We consequently determined the transcriptional profiles of LN GC-Tfh cells following their interactions with LN GC-B cells from HIV-infected and HIV-uninfected individuals, rather than analyzing resting ex-vivo GC-Tfh cells. We observed that proliferating GC-Tfh cells from HIV-infected subjects were transcriptionally different than their HIV-uninfected counterparts, and displayed a significant downregulation of immune- and GC-Tfh-associated pathways and genes. Our results strongly demonstrated that MAF (coding for the transcription factor c-Maf) and its upstream signaling pathway mediators (IL6R and STAT3) were significantly downregulated in HIV-infected subjects, which could contribute to the impaired GC-Tfh and GC-B cell functions reported during infection. We further showed that c-Maf function was associated with the adenosine pathway and that the signaling upstream c-Maf could be partially restored by adenosine deaminase -1 (ADA-1) supplementation. Overall, we identified a novel mechanism that contributes to GC-Tfh cell impairment during HIV infection. Understanding how GC-Tfh cell function is altered in HIV is crucial and could provide critical information about the mechanisms leading to the development and maintenance of effective anti-HIV antibodies.


Subject(s)
HIV Infections/immunology , Proto-Oncogene Proteins c-maf/immunology , T Follicular Helper Cells/immunology , Adult , Chronic Disease , Female , Germinal Center/immunology , Humans , Male , Signal Transduction/immunology
3.
J Allergy Clin Immunol ; 147(4): 1281-1295.e5, 2021 04.
Article in English | MEDLINE | ID: mdl-32905799

ABSTRACT

BACKGROUND: Group 2 innate lymphoid cells (ILC2s) are the dominant innate lymphoid cell population in the lungs at steady state, and their release of type 2 cytokines is a central driver in responding eosinophil infiltration and increased airway hyperreactivity. Our laboratory has identified a unique subset of ILC2s in the lungs that actively produce IL-10 (ILC210s). OBJECTIVE: Our aim was to characterize the effector functions of ILC210s in the development and pathology of allergic asthma. METHODS: IL-4-stimulated ILC210s were isolated to evaluate cytokine secretion, transcription factor signaling, metabolic dependence, and effector functions in vitro. ILC210s were also adoptively transferred into Rag2-/-γc-/- mice, which were then challenged with IL-33 and assessed for airway hyperreactivity and lung inflammation. RESULTS: We have determined that the transcription factors cMaf and Blimp-1 regulate IL-10 expression in ILC210s. Strikingly, our results demonstrate that ILC210s can utilize both autocrine and paracrine signaling to suppress proinflammatory ILC2 effector functions in vitro. Further, this subset dampens airway hyperreactivity and significantly reduces lung inflammation in vivo. Interestingly, ILC210s demonstrated a metabolic dependency on the glycolytic pathway for IL-10 production, shifting from the fatty acid oxidation pathway conventionally utilized for proinflammatory effector functions. CONCLUSION: These findings provide an important and previously unrecognized role of ILC210s in diseases associated with ILC2s such as allergic lung inflammation and asthma. They also provide new insights into the metabolism dependency of proinflammatory and anti-inflammatory ILC2 phenotypes.


Subject(s)
Asthma/immunology , Bronchial Hyperreactivity/immunology , Interleukin-10/immunology , Lymphocytes/immunology , Positive Regulatory Domain I-Binding Factor 1/immunology , Proto-Oncogene Proteins c-maf/immunology , Animals , Bronchoalveolar Lavage Fluid/immunology , Female , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
4.
EMBO J ; 39(13): e104159, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32627520

ABSTRACT

γδ T cells with distinct properties develop in the embryonic and adult thymus and have been identified as critical players in a broad range of infections, antitumor surveillance, autoimmune diseases, and tissue homeostasis. Despite their potential value for immunotherapy, differentiation of γδ T cells in the thymus is incompletely understood. Here, we establish a high-resolution map of γδ T-cell differentiation from the fetal and adult thymus using single-cell RNA sequencing. We reveal novel sub-types of immature and mature γδ T cells and identify an unpolarized thymic population which is expanded in the blood and lymph nodes. Our detailed comparative analysis reveals remarkable similarities between the gene networks active during fetal and adult γδ T-cell differentiation. By performing a combined single-cell analysis of Sox13, Maf, and Rorc knockout mice, we demonstrate sequential activation of these factors during IL-17-producing γδ T-cell (γδT17) differentiation. These findings substantially expand our understanding of γδ T-cell ontogeny in fetal and adult life. Our experimental and computational strategy provides a blueprint for comparing immune cell differentiation across developmental stages.


Subject(s)
Cell Differentiation/immunology , Fetus/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Animals , Autoantigens/genetics , Autoantigens/immunology , Cell Differentiation/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , T-Lymphocytes/cytology
5.
J Clin Invest ; 130(4): 2081-2096, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31945018

ABSTRACT

Macrophages have been linked to tumor initiation, progression, metastasis, and treatment resistance. However, the transcriptional regulation of macrophages driving the protumor function remains elusive. Here, we demonstrate that the transcription factor c-Maf is a critical controller for immunosuppressive macrophage polarization and function in cancer. c-Maf controls many M2-related genes and has direct binding sites within a conserved noncoding sequence of the Csf-1r gene and promotes M2-like macrophage-mediated T cell suppression and tumor progression. c-Maf also serves as a metabolic checkpoint regulating the TCA cycle and UDP-GlcNAc biosynthesis, thus promoting M2-like macrophage polarization and activation. Additionally, c-Maf is highly expressed in tumor-associated macrophages (TAMs) and regulates TAM immunosuppressive function. Deletion of c-Maf specifically in myeloid cells results in reduced tumor burden with enhanced antitumor T cell immunity. Inhibition of c-Maf partly overcomes resistance to anti-PD-1 therapy in a subcutaneous LLC tumor model. Similarly, c-Maf is expressed in human M2 and tumor-infiltrating macrophages/monocytes as well as circulating monocytes of human non-small cell lung carcinoma (NSCLC) patients and critically regulates their immunosuppressive activity. The natural compound ß-glucan downregulates c-Maf expression on macrophages, leading to enhanced antitumor immunity in mice. These findings establish a paradigm for immunosuppressive macrophage polarization and transcriptional regulation by c-Maf and suggest that c-Maf is a potential target for effective tumor immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Immunity, Cellular , Lung Neoplasms/immunology , Macrophage Activation , Macrophages/immunology , Neoplasms, Experimental/immunology , Proto-Oncogene Proteins c-maf/immunology , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/immunology , Humans , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Macrophages/pathology , Male , Mice , Mice, Knockout , Monocytes/immunology , Monocytes/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Proto-Oncogene Proteins c-maf/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology
7.
J Exp Med ; 217(1)2020 01 06.
Article in English | MEDLINE | ID: mdl-31570496

ABSTRACT

CCR6- group 3 innate lymphoid cells (ILC3s) are mediators of intestinal immunity and barrier function that possess the capacity to acquire type 1 effector features and fully convert into ILC1s. The molecular mechanisms governing such plasticity are undefined. Here, we identified c-Maf as an essential regulator of ILC3 homeostasis and plasticity that limits physiological ILC1 conversion. Phenotypic analysis of effector status in Maf-deficient CCR6- ILC3s, coupled with evaluation of global changes in transcriptome, chromatin accessibility, and transcription factor motif enrichment, revealed that c-Maf enforces ILC3 identity. c-Maf promoted ILC3 accessibility and supported RORγt activity and expression of type 3 effector genes. Conversely, c-Maf antagonized type 1 programming, largely through restraint of T-bet expression and function. Mapping of the dynamic changes in chromatin landscape accompanying CCR6- ILC3 development and ILC1 conversion solidified c-Maf as a gatekeeper of type 1 regulatory transformation and a controller of ILC3 fate.


Subject(s)
Immunity, Innate/immunology , Lymphocytes/immunology , Proto-Oncogene Proteins c-maf/immunology , Animals , Cell Lineage/immunology , Chromatin/immunology , Gene Expression Regulation/immunology , Homeostasis/immunology , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Transcription Factors/immunology , Transcriptome/immunology
8.
Sci Rep ; 9(1): 19767, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31875006

ABSTRACT

Autoimmune hemolytic anemia (AIHA) is an acquired autoimmune disease mediated by antibodies against the patient's red blood cells. However, the underlying mechanisms for antibody production are not fully understood. Previous studies of etiology and pathogenesis of AIHA mainly focus on autoreactive B cells that have escaped tolerance mechanisms. Few studies have reported the function of TFH and TFR cells in the process of AIHA. The present study aimed to explore the potential mechanism of TFH and TFR cells in the pathogenesis of AIHA. With the model of murine AIHA, increased ratios of TFH:TFR, elevated serum IL-21 and IL-6 levels, and upregulated Bcl-6 and c-Maf expression were reported. Also, adoptive transfer of purified CD4+CXCR5+CD25- T cells from immunized mice promoted the induction of autoantibody in the AIHA mouse model. Altogether, our data demonstrate the important role of TFH cells for control and induction of AIHA. In the light of the key contributions of TFH cells to the immune response in AIHA, strategies aimed at inhibiting the TFH development or function should be emphasized.


Subject(s)
Anemia, Hemolytic, Autoimmune/immunology , B-Lymphocytes/immunology , Germinal Center/immunology , T-Lymphocytes, Regulatory/immunology , Anemia, Hemolytic, Autoimmune/pathology , Animals , Autoantibodies/immunology , Disease Models, Animal , Germinal Center/pathology , Interleukin-6/immunology , Interleukins/immunology , Mice , Proto-Oncogene Proteins c-bcl-6/immunology , Proto-Oncogene Proteins c-maf/immunology , T-Lymphocytes, Regulatory/pathology
9.
Front Immunol ; 10: 2050, 2019.
Article in English | MEDLINE | ID: mdl-31552021

ABSTRACT

The transcription factor Bach2 is a susceptible gene for numerous autoimmune diseases including systemic lupus erythematosus (SLE). Bach2-/- mice can develop a lupus-like autoimmune disease. However, the exact cellular and molecular mechanisms via which Bach2 protects the hosts from developing autoimmunity remains incompletely understood. Here, we report that Bach2 ablation on T cells, but not B cells, resulted in humoral autoimmunity, and this was associated with expansion of T follicular helper (Tfh) cells and abnormal germinal centers. Bach2 was down-regulated in Tfh cells and directly suppressed by the Tfh-defining transcription factor BCL6. Mechanistically, Bach2 directly suppresses the transcription of Cxcr5 and c-Maf, two key regulators of Tfh cell differentiation. Bach2-deficient Tfh cells were skewed toward the IL-4-producing subset, which induced IgG1 and IgE isotype switching of B cells. Heterozygous Bcl6 deficiency reduced the formation of germinal center and autoantibodies, and ameliorated the pathology in Bach2-deficient mice. Our findings identify Bach2 as a crucial negative regulator of Tfh cells at steady state and prove that Bach2 controls autoimmunity in part by restraining accumulation of pathogenic Tfh cells.


Subject(s)
Autoimmunity/immunology , Basic-Leucine Zipper Transcription Factors/immunology , Interleukin-4/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoantibodies/immunology , Autoimmunity/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Gene Expression Profiling/methods , Germinal Center/immunology , Germinal Center/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/immunology , Proto-Oncogene Proteins c-maf/metabolism , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , Receptors, CXCR5/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism
10.
Immunity ; 51(3): 465-478.e6, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31422869

ABSTRACT

The generation of high-affinity neutralizing antibodies, the objective of most vaccine strategies, occurs in B cells within germinal centers (GCs) and requires rate-limiting "help" from follicular helper CD4+ T (Tfh) cells. Although Tfh differentiation is an attribute of MHC II-restricted CD4+ T cells, the transcription factors driving Tfh differentiation, notably Bcl6, are not restricted to CD4+ T cells. Here, we identified a requirement for the CD4+-specific transcription factor Thpok during Tfh cell differentiation, GC formation, and antibody maturation. Thpok promoted Bcl6 expression and bound to a Thpok-responsive region in the first intron of Bcl6. Thpok also promoted the expression of Bcl6-independent genes, including the transcription factor Maf, which cooperated with Bcl6 to mediate the effect of Thpok on Tfh cell differentiation. Our findings identify a transcriptional program that links the CD4+ lineage with Tfh differentiation, a limiting factor for efficient B cell responses, and suggest avenues to optimize vaccine generation.


Subject(s)
Cell Differentiation/immunology , Proto-Oncogene Proteins c-bcl-6/immunology , Proto-Oncogene Proteins c-maf/immunology , T-Lymphocytes, Helper-Inducer/immunology , Transcription Factors/immunology , Transcription, Genetic/immunology , Animals , Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Female , Gene Expression Regulation/immunology , Germinal Center/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL
11.
Front Immunol ; 10: 421, 2019.
Article in English | MEDLINE | ID: mdl-30941122

ABSTRACT

Mycobacterium tuberculosis (Mtb) can subvert the host defense by skewing macrophage activation toward a less microbicidal alternative activated state to avoid classical effector killing functions. Investigating the molecular basis of this evasion mechanism could uncover potential candidates for host directed therapy against tuberculosis (TB). A limited number of miRNAs have recently been shown to regulate host-mycobacterial interactions. Here, we performed time course kinetics experiments on bone marrow-derived macrophages (BMDMs) and human monocyte-derived macrophages (MDMs) alternatively activated with IL-4, IL-13, or a combination of IL-4/IL-13, followed by infection with Mtb clinical Beijing strain HN878. MiR-143 and miR-365 were highly induced in Mtb-infected M(IL-4/IL-13) BMDMs and MDMs. Knockdown of miR-143 and miR-365 using antagomiRs decreased the intracellular growth of Mtb HN878, reduced the production of IL-6 and CCL5 and promoted the apoptotic death of Mtb HN878-infected M(IL-4/IL-13) BMDMs. Computational target prediction identified c-Maf, Bach-1 and Elmo-1 as potential targets for both miR-143 and miR-365. Functional validation using luciferase assay, RNA-pulldown assay and Western blotting revealed that c-Maf and Bach-1 are directly targeted by miR-143 while c-Maf, Bach-1, and Elmo-1 are direct targets of miR-365. Knockdown of c-Maf using GapmeRs promoted intracellular Mtb growth when compared to control treated M(IL-4/IL-13) macrophages. Meanwhile, the blocking of Bach-1 had no effect and blocking Elmo-1 resulted in decreased Mtb growth. Combination treatment of M(IL-4/IL-13) macrophages with miR-143 mimics or miR-365 mimics and c-Maf, Bach-1, or Elmo-1 gene-specific GapmeRs restored Mtb growth in miR-143 mimic-treated groups and enhanced Mtb growth in miR-365 mimics-treated groups, thus suggesting the Mtb growth-promoting activities of miR-143 and miR-365 are mediated at least partially through interaction with c-Maf, Bach-1, and Elmo-1. We further show that knockdown of miR-143 and miR-365 in M(IL-4/IL-13) BMDMs decreased the expression of HO-1 and IL-10 which are known targets of Bach-1 and c-Maf, respectively, with Mtb growth-promoting activities in macrophages. Altogether, our work reports a host detrimental role of miR-143 and miR-365 during Mtb infection and highlights for the first time the role and miRNA-mediated regulation of c-Maf, Bach-1, and Elmo-1 in Mtb-infected M(IL-4/IL-13) macrophages.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Basic-Leucine Zipper Transcription Factors/immunology , Macrophages/microbiology , MicroRNAs/immunology , Mycobacterium tuberculosis/growth & development , Proto-Oncogene Proteins c-maf/immunology , Animals , Interleukin-13/pharmacology , Interleukin-4/pharmacology , Macrophages/drug effects , Macrophages/immunology , Male , Mice, Inbred BALB C , Tuberculosis/genetics , Tuberculosis/immunology , Tuberculosis/microbiology
12.
Sci Rep ; 9(1): 6135, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30992496

ABSTRACT

The maintenance of homeostasis in the gut is a major challenge for the immune system. Here we demonstrate that the transcription factor MAF plays a central role in T cells for the prevention of gastro-intestinal inflammation. Conditional knock out mice lacking Maf in all T cells developed spontaneous late-onset colitis, correlating with a decrease of FOXP3+RORγt+ T cells proportion, dampened IL-10 production in the colon and an increase of inflammatory TH17 cells. Strikingly, FOXP3+ specific conditional knock out mice for MAF did not develop colitis and demonstrated normal levels of IL-10 in their colon, despite the incapacity of regulatory T cells lacking MAF to suppress colon inflammation in Rag1-/- mice transferred with naïve CD4+ T cells. We showed that one of the cellular sources of IL-10 in the colon of these mice are TH17 cells. Thus, MAF is critically involved in the maintenance of the gut homeostasis by regulating the balance between Treg and TH17 cells either at the level of their differentiation or through the modulation of their functions.


Subject(s)
Colitis/genetics , Proto-Oncogene Proteins c-maf/genetics , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology , Animals , Cells, Cultured , Colitis/immunology , Colitis/pathology , Female , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/immunology , Gene Deletion , Interleukin-10/analysis , Interleukin-10/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/analysis , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Proto-Oncogene Proteins c-maf/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
13.
Nat Immunol ; 19(10): 1126-1136, 2018 10.
Article in English | MEDLINE | ID: mdl-30201991

ABSTRACT

Different types of effector and memory T lymphocytes are induced and maintained in protective or pathological immune responses. Here we characterized two human CD4+ TH17 helper cell subsets that, in the recently activated state, could be distinguished on the basis of their expression of the anti-inflammatory cytokine IL-10. IL-10+ TH17 cells upregulated a variety of genes encoding immunoregulatory molecules, as well as genes whose expression is characteristic of tissue-resident T cells. In contrast, IL-10- TH17 cells maintained a pro-inflammatory gene-expression profile and upregulated the expression of homing receptors that guide recirculation from tissues to blood. Expression of the transcription factor c-MAF was selectively upregulated in IL-10+ TH17 cells, and it was bound to a large set of enhancer-like regions and modulated the immunoregulatory and tissue-residency program. Our results identify c-MAF as a relevant factor that drives two highly divergent post-activation fates of human TH17 cells and provide a framework with which to investigate the role of these cells in physiology and immunopathology.


Subject(s)
Interleukin-10/immunology , Proto-Oncogene Proteins c-maf/immunology , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology , Chemotaxis, Leukocyte/immunology , Gene Expression Regulation/immunology , Humans , Inflammation/immunology , Interleukin-10/biosynthesis , Proto-Oncogene Proteins c-maf/metabolism , T-Lymphocyte Subsets/metabolism , Th17 Cells/metabolism
14.
Nat Immunol ; 19(5): 497-507, 2018 05.
Article in English | MEDLINE | ID: mdl-29662170

ABSTRACT

The transcription factor c-Maf induces the anti-inflammatory cytokine IL-10 in CD4+ T cells in vitro. However, the global effects of c-Maf on diverse immune responses in vivo are unknown. Here we found that c-Maf regulated IL-10 production in CD4+ T cells in disease models involving the TH1 subset of helper T cells (malaria), TH2 cells (allergy) and TH17 cells (autoimmunity) in vivo. Although mice with c-Maf deficiency targeted to T cells showed greater pathology in TH1 and TH2 responses, TH17 cell-mediated pathology was reduced in this context, with an accompanying decrease in TH17 cells and increase in Foxp3+ regulatory T cells. Bivariate genomic footprinting elucidated the c-Maf transcription-factor network, including enhanced activity of NFAT; this led to the identification and validation of c-Maf as a negative regulator of IL-2. The decreased expression of the gene encoding the transcription factor RORγt (Rorc) that resulted from c-Maf deficiency was dependent on IL-2, which explained the in vivo observations. Thus, c-Maf is a positive and negative regulator of the expression of cytokine-encoding genes, with context-specific effects that allow each immune response to occur in a controlled yet effective manner.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Gene Regulatory Networks/immunology , Interleukin-2/biosynthesis , Proto-Oncogene Proteins c-maf/immunology , Animals , Interleukin-2/immunology , Mice
15.
Int J Mol Med ; 41(5): 2935-2942, 2018 May.
Article in English | MEDLINE | ID: mdl-29484383

ABSTRACT

cMaf is a leucine-zipper transcription factor that is involved in cell differentiation, oncogenic transformation, and human diseases; however, the functions of cMaf in inflammatory responses in macrophages are still not fully understood. Western blot analysis showed that cMaf expression was induced by lipopolysaccharide (LPS) stimulation in mouse macrophages. An enzyme-linked immunosorbent assay was performed to detect the level of expression of inflammatory cytokines after knockdown of cMaf expression in macrophages using a small interfering RNA (siRNA). Signaling pathway inhibitor analyses indicated that extracellular signal-related kinase and phosphoinositide 3-kinase contribute to mammalian target of rapamycin phosphorylation (mTOR), which controls cMaf expression at the translational level by regulating the expression of eIF4E-binding protein 1 and S6 ribosomal kinase 1 in response to Toll-like receptor 4 signaling. Histopathological findings of the lung and a survival analysis showed that mice transplanted with cMaf-knockdown macrophages were more susceptible to LPS challenge. Taken together, our study revealed that the control of cMaf expression at the translational level by mTOR regulated the expression of inflammatory genes in response to LPS challenge. Moreover, cMaf protected mice from septic shock indicating that cMaf may improve host fitness, thereby enabling the survival of certain infectious diseases.


Subject(s)
Inflammation/immunology , Lipopolysaccharides/immunology , Macrophages/immunology , Proto-Oncogene Proteins c-maf/immunology , Signal Transduction , TOR Serine-Threonine Kinases/immunology , Toll-Like Receptor 4/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation , Gene Knockdown Techniques , Inflammation/genetics , MAP Kinase Signaling System , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-maf/genetics , RAW 264.7 Cells , TOR Serine-Threonine Kinases/genetics , Toll-Like Receptor 4/genetics
16.
Immunobiology ; 221(1): 48-55, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26307002

ABSTRACT

Macrophages function as an essential component of innate immune system, contributing to both the initiation and appropriate resolution of inflammation. The exposure of macrophages to the microbial products, such as lipopolysaccharide (LPS), can strongly shift the balance between tissue homeostasis and inflammation in favor of causing systemic damage, in which macrophage M1 polarization play important roles. Strategies aiming at restoring the balance of macrophage polarization remain to be further explored. Herein, we have demonstrated that poliovirus receptor (PVR), the receptor of TIGIT, was dramatically upregulated on the surface of mouse peritoneal macrophages when exposed to LPS. TIGIT-Fc fusion protein not only inhibited the macrophage activation, but also skewed M1/M2 balance toward an anti-inflammatory profile, especially enhanced the secretion of IL-10. The activation of TIGIT/PVR pathway in macrophages correlated with increased nuclear translocation of c-Maf, which promotes IL-10 transcription. Treatment with fibroblasts stably secreting TIGIT-Fc fusion protein significantly reversed the lethal and sublethal endotoxic shock, which facilitated peritoneal macrophages to switch towards anti-inflammatory M2 cytokine profiles. These findings highlight a novel role of the TIGIT/PVR pathway in macrophage M2 polarization and suggest that TIGIT may have the potential to optimize the treatment of macrophage-involved inflammatory diseases.


Subject(s)
Macrophages, Peritoneal/drug effects , Receptors, Immunologic/immunology , Receptors, Virus/immunology , Recombinant Fusion Proteins/pharmacology , Animals , Gene Expression Regulation , HEK293 Cells , Humans , Immunoglobulin Fc Fragments/genetics , Inflammation/chemically induced , Inflammation/immunology , Inflammation/mortality , Inflammation/pathology , Interleukin-10/genetics , Interleukin-10/immunology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Phenotype , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/immunology , Receptors, Immunologic/genetics , Receptors, Virus/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction , Survival Analysis
17.
J Immunol ; 195(2): 488-97, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26056255

ABSTRACT

The two best-characterized types of CD4(+) regulatory T cells (Tregs) are Foxp3(+) Tregs and Foxp3(-) type 1 regulatory (Tr1) cells. The ability of Foxp3(+) Tregs and Tr1 cells to suppress adaptive immune responses is well known, but how these cells regulate innate immunity is less defined. We discovered that CD44(hi)Foxp3(-) T cells from unmanipulated mice are enriched in Tr1 cell precursors, enabling differentiation of cells that express IL-10, as well as Tr1-associated cell surface markers, CD49b and LAG-3, and transcription factors, cMaf, Blimp-1, and AhR. We compared the ability of Tr1 cells versus Foxp3(+) Tregs to suppress IL-1ß production from macrophages following LPS and ATP stimulation. Surprisingly, Tr1 cells, but not Foxp3(+) Tregs, inhibited the transcription of pro-IL-1ß mRNA, inflammasome-mediated activation of caspase-1, and secretion of mature IL-1ß. Consistent with the role for IL-10 in Tr1 cell-mediated suppression, inhibition of inflammasome activation and IL-1ß secretion was abrogated in IL-10R-deficient macrophages. Moreover, IL-1ß production from macrophages derived from Nlrp3(A350V) knockin mice, which carry a mutation found in cryopyrin-associated periodic syndrome patients, was suppressed by Tr1 cells but not Foxp3(+) Tregs. Using an adoptive transfer model, we found a direct correlation between Tr1 cell engraftment and protection from weight loss in mice expressing a gain-of-function NLRP3. Collectively, these data provide the first evidence for a differential role of Tr1 cells and Foxp3(+) Tregs in regulating innate immune responses. Through their capacity to produce high amounts of IL-10, Tr1 cells may have unique therapeutic effects in disease-associated inflammasome activation.


Subject(s)
Forkhead Transcription Factors/immunology , Inflammasomes/immunology , Interleukin-10/immunology , T-Lymphocytes, Regulatory/immunology , Adenosine Triphosphate/pharmacology , Adoptive Transfer , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Differentiation , Coculture Techniques , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Inflammasomes/drug effects , Inflammasomes/genetics , Integrin alpha2/genetics , Integrin alpha2/immunology , Interleukin-10/genetics , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/immunology , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunology , Signal Transduction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/transplantation , Transcription Factors/genetics , Transcription Factors/immunology , Lymphocyte Activation Gene 3 Protein
18.
J Immunol ; 195(1): 217-26, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26019273

ABSTRACT

Proper regulation of microbial-induced cytokines is critical to intestinal immune homeostasis. Acute stimulation of nucleotide-binding oligomerization domain 2 (NOD2), the Crohn's disease-associated sensor of bacterial peptidoglycan, induces cytokines. However, chronic NOD2 stimulation in macrophages decreases cytokines upon pattern recognition receptor (PRR) restimulation; cytokine attenuation to PRR stimulation is similarly observed in intestinal macrophages. The role for the transcriptional repressors Twist1 and Twist2 in regulating PRR-induced cytokine outcomes is poorly understood and has not been reported for NOD2. We found that Twist1 and Twist2 were required for optimal cytokine downregulation during acute and, particularly, chronic NOD2 stimulation of human macrophages. Consistently, Twist1 and Twist2 expression was increased after chronic NOD2 stimulation; this increased expression was IL-10 and TGF-ß dependent. Although Twist1 and Twist2 did not coregulate each other's expression, they cooperated to enhance binding to cytokine promoters after chronic NOD2 stimulation. Moreover, Twist1 and Twist2 contributed to enhance expression and promoter binding of the proinflammatory inhibitor c-Maf and the transcriptional repressor Bmi1. Restoring c-Maf and Bmi1 expression in Twist-deficient macrophages restored NOD2-induced cytokine downregulation. Furthermore, with chronic NOD2 stimulation, Twist1 and Twist2 contributed to the decreased expression and cytokine promoter binding of the transcriptional activators activating transcription factor 4, C/EBPα, Runx1, and Runx2. Knockdown of these transcriptional activators in Twist-deficient macrophages restored cytokine downregulation after chronic NOD2 stimulation. Finally, NOD2 synergized with additional PRRs to increase Twist1 and Twist2 expression and Twist-dependent pathways. Therefore, after chronic NOD2 stimulation Twist1 and Twist2 coordinate the regulation of both transcriptional activators and repressors, thereby mediating optimal cytokine downregulation.


Subject(s)
Macrophage Activation/drug effects , Macrophages/immunology , Nod2 Signaling Adaptor Protein/immunology , Nuclear Proteins/immunology , Repressor Proteins/immunology , Twist-Related Protein 1/immunology , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Activating Transcription Factor 4/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/immunology , Antibodies, Neutralizing/pharmacology , CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/immunology , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/immunology , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/immunology , Gene Expression Regulation , Humans , Interleukin-10/antagonists & inhibitors , Interleukin-10/genetics , Interleukin-10/immunology , Macrophages/cytology , Macrophages/drug effects , Nod2 Signaling Adaptor Protein/agonists , Nod2 Signaling Adaptor Protein/genetics , Nuclear Proteins/genetics , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/immunology , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Repressor Proteins/genetics , Signal Transduction , Transcription, Genetic , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Twist-Related Protein 1/genetics
19.
Nat Commun ; 6: 6702, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25850032

ABSTRACT

Decline in immune function and inflammation concomitantly develop with ageing. Here we focus on the impact of this inflammatory environment on T cells, and demonstrate that in contrast to successful tumour elimination in young mice, replenishment of tumour-specific CD4(+) T cells fails to induce tumour regression in aged hosts. The impaired antitumour effect of CD4(+) T cells with their defective Th1 differentiation in an aged environment is restored by interleukin (IL)-6 blockade or IL-6 deficiency. IL-6 blockade also restores the impaired ability of CD4(+) T cells to promote CD8(+) T-cell-dependent tumour elimination in aged mice, which requires IFN-γ. Furthermore, IL-6-stimulated production of IL-4/IL-21 through c-Maf induction is responsible for impaired Th1 differentiation. IL-6 also contributes to IL-10 production from CD4(+) T cells in aged mice, causing attenuated responses of CD8(+) T cells. These findings suggest that IL-6 serves as an extrinsic factor counteracting CD4(+) T-cell-mediated immunity against tumour in old age.


Subject(s)
Aging/immunology , Cell Differentiation/immunology , Fibrosarcoma/immunology , Interleukin-6/immunology , Th1 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular/immunology , Interferon-gamma/immunology , Interleukin-10/immunology , Interleukin-4/immunology , Interleukins/immunology , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Proto-Oncogene Proteins c-maf/immunology , Tumor Escape
20.
Int Immunol ; 27(8): 405-15, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25862525

ABSTRACT

Aryl hydrocarbon receptor (Ahr), a transcription factor, plays a critical role in autoimmune inflammation of the intestine. In addition, microRNAs (miRNAs), small non-coding oligonucleotides, mediate pathogenesis of inflammatory bowel diseases (IBD). However, the precise mechanism and interactions of these molecules in IBD pathogenesis have not yet been investigated. We analyzed the role of Ahr and Ahr-regulated miRNAs in colonic inflammation. Our results show that deficiency of Ahr in intestinal epithelial cells in mice exacerbated inflammation in dextran sodium sulfate-induced colitis. Deletion of Ahr in T cells attenuated colitis, which was manifested by suppressed Th17 cell infiltration into the lamina propria. Candidate miRNA analysis showed that induction of colitis elevated expression of the miR-212/132 cluster in the colon of wild-type mice, whereas in Ahr (-/-) mice, expression was clearly lower. Furthermore, miR-212/132(-/-) mice were highly resistant to colitis and had reduced levels of Th17 cells and elevated levels of IL-10-producing CD4(+) cells. In vitro analyses revealed that induction of type 1 regulatory T (Tr1) cells was significantly elevated in miR-212/132(-/-) T cells with increased c-Maf expression. Our findings emphasize the vital role of Ahr in intestinal homeostasis and suggest that inhibition of miR-212/132 represents a viable therapeutic strategy for treating colitis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Colitis/genetics , Interleukin-10/genetics , MicroRNAs/genetics , Receptors, Aryl Hydrocarbon/genetics , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/immunology , Cell Proliferation , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Dextran Sulfate , Female , Gene Expression Regulation , Homeostasis/immunology , Interleukin-10/immunology , Intestines/immunology , Intestines/pathology , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/immunology , Molecular Sequence Data , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/immunology , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/immunology , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/immunology , Th17 Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL