Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Immunol ; 207(1): 55-64, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34135066

ABSTRACT

Effector CD4+ T lymphocytes contribute to inflammation and tissue damage in psoriasis, but the underlying molecular mechanisms remain poorly understood. The transcription factor CREMα controls effector T cell function in people with systemic autoimmune diseases. The inhibitory surface coreceptor PD-1 plays a key role in the control of effector T cell function and its therapeutic inhibition in patients with cancer can cause psoriasis. In this study, we show that CD4+ T cells from patients with psoriasis and psoriatic arthritis exhibit increased production of IL-17 but decreased expression of IL-2 and PD-1. In genetically modified mice and Jurkat T cells CREMα expression was linked to low PD-1 levels. We demonstrate that CREMα is recruited to the proximal promoter of PDCD1 in which it trans-represses gene expression and corecruits DNMT3a-mediating DNA methylation. As keratinocytes limit inflammation by PD-1 ligand expression and, in this study, reported reduced expression of PD-1 on CD4+ T cells is linked to low IL-2 and high IL-17A production, our studies reveal a molecular pathway in T cells from people with psoriasis that can deserve clinical exploitation.


Subject(s)
Arthritis, Psoriatic/immunology , CD4-Positive T-Lymphocytes/immunology , Cyclic AMP Response Element Modulator/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Humans , Mice , Mice, Inbred C57BL
2.
Clin Nutr ; 40(5): 3263-3278, 2021 05.
Article in English | MEDLINE | ID: mdl-33183881

ABSTRACT

BACKGROUND & AIMS: The micronutrient zinc is essential for proper immune function. Consequently, zinc deficiency leads to impaired immune function, as seen in decreased secretion of interleukin (IL)-2 by T cells. Although this association has been known since the late 1980s, the underlying molecular mechanisms are still unknown. Zinc deficiency and reduced IL-2 levels are especially found in the elderly, which in turn are prone to chronic diseases. Here, we describe a new molecular link between zinc deficiency and reduced IL-2 expression in T cells. METHODS: The effects of zinc deficiency were first investigated in vitro in the human T cell lines Jurkat and Hut-78 and complemented by in vivo data from zinc-supplemented pigs. A short- and long-term model for zinc deficiency was established. Zinc levels were detected by flow cytometry and expression profiles were investigated on the mRNA and protein level. RESULTS: The expression of the transcription factor cAMP-responsive-element modulator α (CREMα) is increased during zinc deficiency in vitro, due to increased protein phosphatase 2A (PP2A) activity, resulting in decreased IL-2 production. Additionally, zinc supplementation in vivo reduced CREMα levels causing increased IL-2 expression. On epigenetic levels increased CREMα binding to the IL-2 promoter is mediated by histone deacetylase 1 (HDAC1). The HDAC1 activity is inhibited by zinc. Moreover, deacetylation of the activating histone mark H3K9 was increased under zinc deficiency, resulting in reduced IL-2 expression. CONCLUSIONS: With the transcription factor CREMα a molecular link was uncovered, connecting zinc deficiency with reduced IL-2 production due to enhanced PP2A and HDAC1 activity.


Subject(s)
Cyclic AMP Response Element Modulator/immunology , Gene Expression/genetics , Gene Silencing , Interleukin-2/biosynthesis , T-Lymphocytes/immunology , Zinc/deficiency , Zinc/immunology , Animals , Cyclic AMP Response Element Modulator/genetics , Disease Models, Animal , Gene Expression/immunology , Humans , In Vitro Techniques , Interleukin-2/genetics , Interleukin-2/immunology , Swine
3.
Cell Physiol Biochem ; 52(3): 580-594, 2019.
Article in English | MEDLINE | ID: mdl-30897323

ABSTRACT

BACKGROUND/AIMS: Studies have indicated that sympathetic activity enhances GLUT4 expression (Slc2a4 gene) by activating beta-adrenergic receptors. This could be mediated by a direct enhancer effect of cyclic AMP-responsive element binding protein (CREB) and family members upon Slc2a4 gene. However, a cAMP responsive element (CRE) in Slc2a4 promoter has never been demonstrated. METHODS: Slc2a4 CRE-site was searched by in silico analysis. In skeletal muscles from rats displaying high sympathetic activity (SHR), Slc2a4 CRE-site was investigated by electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assay; and Slc2a4 expression was analyzed by RT-qPCR. Functional activity of the CRE-site was investigated by luciferase assay, 2 hours after 8-br-cAMP stimulation, in 3T3L1 adipocytes transientely transfected with native and mutated CRE-sites. RESULTS: In silico analysis indicated the -480/-473 segment as a putative CRE-site, with 62.5% of identity to CRE consensus sequence, and highly preserved in mouse, rat and human. CREB/CREM binding in this CRE-site was confirmed to occur in vitro (EMSA) and in vivo (ChIP assay). Enhancer activity of this segment in Slc2a4 transcription was confirmed in 3T3-L1 cells. Finally, in extensor digitorum longus muscle from SHR, 80% increase in Slc2a4 mRNA expression was observed to be accompanied by increased CREB/CREM binding into the CRE-site both in vitro and in vivo. CONCLUSION: This study demonstrates the presence of a functional CRE-site at -480/-473 sequence of the Slc2a4 gene. This CRE-site has an enhancing activity on Slc2a4 expression and participates in the Slc2a4 increased expression observed in glycolytic muscles of rats displaying high sympathetic activity.


Subject(s)
Cyclic AMP Response Element Modulator/metabolism , Glucose Transporter Type 4/metabolism , 3T3-L1 Cells , 5' Untranslated Regions , Animals , Base Sequence , Cyclic AMP/metabolism , Cyclic AMP Response Element Modulator/immunology , Electrophoretic Mobility Shift Assay , Glucose Transporter Type 4/genetics , Male , Mice , Muscle, Skeletal/metabolism , Mutagenesis , Promoter Regions, Genetic , Protein Binding , Rats , Rats, Inbred SHR , Rats, Wistar , Transcriptional Activation
4.
Proc Natl Acad Sci U S A ; 115(37): 9288-9293, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150402

ABSTRACT

Th17 cells favor glycolytic metabolism, and pyruvate dehydrogenase (PDH) is the key bifurcation enzyme, which in its active dephosphorylated form advances the oxidative phosphorylation from glycolytic pathway. The transcriptional factor, inducible cAMP early repressor/cAMP response element modulator (ICER/CREM), has been shown to be induced in Th17 cells and to be overexpressed in CD4+ T cells from the patients with systemic lupus erythematosus (SLE). We found that glycolysis and lactate production in in vitro Th17-polarized T cells was reduced and that the expression of pyruvate dehydrogenase phosphatase catalytic subunit 2 (PDP2), an enzyme that converts the inactive PDH to its active form, and PDH enzyme activity were increased in Th17 cells from ICER/CREM-deficient animals. ICER was found to bind to the Pdp2 promoter and suppress its expression. Furthermore, forced expression of PDP2 in CD4+ cells reduced the in vitro Th17 differentiation, whereas shRNA-based suppression of PDP2 expression increased in vitro Th17 differentiation and augmented experimental autoimmune encephalomyelitis. At the translational level, PDP2 expression was decreased in memory Th17 cells from patients with SLE and forced expression of PDP2 in CD4+ T cells from lupus-prone MRL/lpr mice and patients with SLE suppressed Th17 differentiation. These data demonstrate the direct control of energy production during Th17 differentiation in health and disease by the transcription factor ICER/CREM at the PDH metabolism bifurcation level.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Enzymologic , Phosphoprotein Phosphatases/biosynthesis , Response Elements , Th17 Cells/enzymology , Animals , Catalytic Domain , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/immunology , Cyclic AMP Response Element Modulator/metabolism , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Male , Mice , Mice, Knockout , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/immunology , Th17 Cells/immunology , Th17 Cells/pathology
5.
Cell Rep ; 21(1): 154-167, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978469

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) and dendritic cells (DCs) arise from common progenitors. Tumor-derived factors redirect differentiation from immune-promoting DCs to tolerogenic MDSCs, an immunological hallmark of cancer. Indeed, in vitro differentiation of DCs from human primary monocytes results in the generation of MDSCs under tumor-associated conditions (PGE2 or tumor cell-conditioned media). Comparison of MDSC and DC DNA methylomes now reveals extensive demethylation with specific gains of DNA methylation and repression of immunogenic-associated genes occurring in MDSCs specifically, concomitant with increased DNA methyltransferase 3A (DNMT3A) levels. DNMT3A downregulation erases MDSC-specific hypermethylation, and it abolishes their immunosuppressive capacity. Primary MDSCs isolated from ovarian cancer patients display a similar hypermethylation signature in connection with PGE2-dependent DNMT3A overexpression. Our study links PGE2- and DNMT3A-dependent hypermethylation with immunosuppressive MDSC functions, providing a promising target for therapeutic intervention.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Dinoprostone/pharmacology , Gene Expression Regulation, Neoplastic , Immune Tolerance , Myeloid-Derived Suppressor Cells/drug effects , Ovarian Neoplasms/genetics , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Lineage/immunology , Chemokine CCL22/genetics , Chemokine CCL22/immunology , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/immunology , Culture Media, Conditioned/pharmacology , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/immunology , DNA (Cytosine-5-)-Methyltransferases/immunology , DNA Methylation , DNA Methyltransferase 3A , Female , Humans , Monocytes/drug effects , Monocytes/immunology , Multigene Family , Myeloid-Derived Suppressor Cells/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Primary Cell Culture
6.
Beijing Da Xue Xue Bao Yi Xue Ban ; 48(6): 1100-1104, 2016 12 18.
Article in Chinese | MEDLINE | ID: mdl-27987522

ABSTRACT

Systemic lupus erythematosus (SLE) is a prototypic systemic autoimmune disease, which characterized by complex immunological abnormalities and multiple tissue and organ damages. The etiology and pathogenesis of SLE have not been entirely recognized. Genetic, environmental and viral infections and other factors might be related to the pathogenetic mechanisms of SLE. Interleukin-2 (IL-2) is a critical cytokine produced by T cells upon activation and is important for the generation of T regulatory cells and activation-induced cell death. In SLE patients, T cells display decreased capacity to produce IL-2. Impaired IL-2 expression resulted in decreased generation of regulatory T lymphocytes, and defect of activation-induced cell death. Former researches indicated that IL-2 deficiency in SLE is important for the pathogenesis and treatment of SLE. Several regulating molecules can affect the transcription of IL-2 gene and had an important role in the pathogenesis of SLE. These molecules include cyclic AMP-responsive element modulator (CREM), protein phosphatase 2A (PP2A), E-74 like factor 1 (Elf-1), B lymphocyte induced maturation protein-1 (Blimp-1) and interferon regulator factor 5 (IRF-5). CREM is a transcriptional inhibitor that can repress the transcription of the IL-2 gene by binding to the promoter of the IL-2 gene. PP2A is a Ser/Thr phosphatase that expressed in eukaryotic cells ubiquitously, it represents a negative regulator of the IL-2 gene promoter activity. Elf-1 belongs to the Ets family of transcription factors and can promote the expression of IL-2. Blimp-1 is a crucial transcription factors for regulating B lymphocyte terminal differentiation, an important function of Blimp-1 in T cells is to repress IL-2 gene transcription directly. Interferon regulatory factors (IRFs) are distinctive transcriptional regulators of type I interferons (IFNs) and IFN inducible genes, IRF-5 is a member of the IRFs family. IRF-5 is found to be increased in SLE and can regulate the production of IL-2 negatively. PP2A can inhibit the synthesis of IL-2 in two ways: on the one hand, activating the IL-2 transcription inhibitory factor CREMα, on the other hand, inhibiting IL-2 stimulating transcription factor Elf-1. While IRF-5 can activate the IL-2 transcription negative regulator Blimp-1 as to inhibit IL-2 expression. These molecules participate in the regulation of IL-2 through different pathways. This paper reviews the current knowledge of IL-2 signaling pathway regulating molecules in SLE.


Subject(s)
Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/physiopathology , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/immunology , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , Protein Phosphatase 2/genetics , Protein Phosphatase 2/immunology , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Transcription Factors/genetics , Transcription Factors/immunology
7.
Proc Natl Acad Sci U S A ; 113(46): E7231-E7239, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27799548

ABSTRACT

The protein encoded by the autoimmune-associated protein tyrosine phosphatase nonreceptor type 22 gene, PTPN22, has wide-ranging effects in immune cells including suppression of T-cell receptor signaling and promoting efficient production of type I interferons (IFN-I) by myeloid cells. Here we show that mice deficient in PTPN22 resist chronic viral infection with lymphocytic choriomeningitis virus clone 13 (LCMV cl13). The numbers and function of viral-specific CD4 T lymphocytes is greatly enhanced, whereas expression of the IFNß-induced IL-2 repressor, cAMP-responsive element modulator (CREM) is reduced. Reduction of CREM expression in wild-type CD4 T lymphocytes prevents the loss of IL-2 production by CD4 T lymphocytes during infection with LCMV cl13. These findings implicate the IFNß/CREM/IL-2 axis in regulating T-lymphocyte function during chronic viral infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocytic Choriomeningitis/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , Animals , Cyclic AMP Response Element Modulator/immunology , Interferon Type I/immunology , Interleukin-10/immunology , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics
8.
Oncotarget ; 6(36): 38538-51, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26459392

ABSTRACT

A characteristic feature of allergic diseases is the appearance of a subset of CD4+ cells known as TH2 cells, which is controlled by transcriptional and epigenetic mechanisms. We aimed to analyze the role of CREM, a known transcriptional activator of T cells, with regard to TH2 responses and allergic diseases in men and mice. Here we demonstrate that T cells of asthmatic children and PBMCs of adults with atopy express lower mRNA levels of the transcription factor CREM compared to cells from healthy controls. CREM deficiency in murine T cells results in enhanced TH2 effector cytokines in vitro and in vivo and CREM-/- mice demonstrate stronger airway hyperresponsiveness in an OVA-induced asthma model. Mechanistically, both direct CREM binding to the IL-4 and IL-13 promoter as well as a decreased IL-2 dependent STAT5 activation suppress the TH2 response. Accordingly, mice selectively overexpressing CREMα in T cells display decreased TH2 type cytokines in vivo and in vitro, and are protected in an asthma model. Thus, we provide evidence that CREM is a negative regulator of the TH2 response and determines the outcome of allergic asthma.


Subject(s)
Cyclic AMP Response Element Modulator/immunology , Inflammation/immunology , Th2 Cells/immunology , Adolescent , Adult , Animals , Asthma/immunology , Child , Cyclic AMP Response Element Modulator/biosynthesis , Cytokines/immunology , Down-Regulation , Female , Humans , Hypersensitivity/immunology , Male , Mice , Mice, Transgenic , Young Adult
9.
J Clin Invest ; 124(5): 2234-45, 2014 May.
Article in English | MEDLINE | ID: mdl-24667640

ABSTRACT

Tissue inflammation in several autoimmune diseases, including SLE and MS, has been linked to an imbalance of IL-17-producing Th (Th17) cells and Tregs; however, the factors that promote Th17-driven autoimmunity are unclear. Here, we present evidence that the calcium/calmodulin-dependent protein kinase IV (CaMK4) is increased and required during Th17 cell differentiation. Isolation of naive T cells from a murine model of lupus revealed increased levels of CaMK4 following stimulation with Th17-inducing cytokines but not following Treg, Th1, or Th2 induction. Furthermore, naive T cells from mice lacking CaMK4 did not produce IL-17. Genetic or pharmacologic inhibition of CaMK4 decreased the frequency of IL-17-producing T cells and ameliorated EAE and lupus-like disease in murine models. Inhibition of CaMK4 reduced Il17 transcription through decreased activation of the cAMP response element modulator α (CREM-α) and reduced activation of the AKT/mTOR pathway, which is known to enhance Th17 differentiation. Importantly, silencing CaMK4 in T cells from patients with SLE and healthy individuals inhibited Th17 differentiation through reduction of IL17A and IL17F mRNA. Collectively, our results suggest that CaMK4 inhibition has potential as a therapeutic strategy for Th17-driven autoimmune diseases.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/immunology , Cyclic AMP Response Element Modulator/immunology , Lupus Erythematosus, Systemic/immunology , Multiple Sclerosis/immunology , Proto-Oncogene Proteins c-akt/immunology , TOR Serine-Threonine Kinases/immunology , Th17 Cells/immunology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 4/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cyclic AMP Response Element Modulator/genetics , Enzyme Activation/genetics , Enzyme Activation/immunology , Female , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/therapy , Male , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology , TOR Serine-Threonine Kinases/genetics , Th17 Cells/pathology
10.
Amino Acids ; 45(6): 1373-83, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24100545

ABSTRACT

Human cyclic AMP response modulator proteins (CREMs) are encoded by the CREM gene, which generates 30 or more different CREM protein isoforms. They are members of the leucine zipper protein superfamily of nuclear transcription factors. CREM proteins are known to be implicated in a plethora of important cellular processes within the CNS. Amazingly, little is known about their cellular and regional distribution in the brain, however. Therefore, we studied by means of immunohistochemistry and Western blotting the expression patterns of CREM in developing and adult human brain, as well as in brains of Alzheimer's disease patients. CREM immunoreactivity was found to be widely but unevenly distributed in the adult human brain. Its localization was confined to neurons. In immature human brains, CREM multiple neuroblasts and radial glia cells expressed CREM. In Alzheimer's brain, we found an increased cellular expression of CREM in dentate gyrus neurons as compared to controls. We discuss our results with regard to the putative roles of CREM in brain development and in cognition.


Subject(s)
Aging , Alzheimer Disease/metabolism , Cyclic AMP Response Element Modulator/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/pathology , Neurons/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Cyclic AMP Response Element Modulator/biosynthesis , Cyclic AMP Response Element Modulator/immunology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neurons/cytology , Neurons/pathology , Rats , Rats, Sprague-Dawley
11.
J Biol Chem ; 288(44): 31880-7, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24047902

ABSTRACT

T cell receptor-αß(+) CD3(+)CD4(-)CD8(-) "double-negative" T cells are expanded in the peripheral blood of patients with systemic lupus erythematosus and autoimmune lymphoproliferative syndrome. In both disorders, double-negative T cells infiltrate tissues, induce immunoglobulin production, and secrete proinflammatory cytokines. Double-negative T cells derive from CD8(+) T cells through down-regulation of CD8 surface co-receptors. However, the molecular mechanisms orchestrating this process remain unclear. Here, we demonstrate that the transcription factor cAMP-responsive element modulator α (CREMα), which is expressed at increased levels in T cells from systemic lupus erythematosus patients, contributes to transcriptional silencing of CD8A and CD8B. We provide the first evidence that CREMα trans-represses a regulatory element 5' of the CD8B gene. Therefore, CREMα represents a promising candidate in the search for biomarkers and treatment options in diseases in which double-negative T cells contribute to the pathogenesis.


Subject(s)
CD8 Antigens/biosynthesis , CD8-Positive T-Lymphocytes/metabolism , Cyclic AMP Response Element Modulator/metabolism , Gene Silencing , Lupus Erythematosus, Systemic/metabolism , Repressor Proteins/metabolism , Response Elements , CD3 Complex/genetics , CD3 Complex/immunology , CD3 Complex/metabolism , CD4 Antigens/genetics , CD4 Antigens/immunology , CD4 Antigens/metabolism , CD8 Antigens/genetics , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/immunology , Female , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Male , Repressor Proteins/genetics , Repressor Proteins/immunology , Transcription, Genetic/genetics , Transcription, Genetic/immunology
12.
Biochem J ; 456(3): 463-73, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24007532

ABSTRACT

cAMP signalling is both a major pathway as well as a key therapeutic target for inducing immune tolerance and is involved in Treg cell (regulatory T-cell) function. To achieve potent immunoregulation, cAMP can act through several downstream effectors. One proposed mechanism is that cAMP-mediated suppression, including immunosuppression by Treg cells, results from activation of PKA (protein kinase A) leading to the induction of the transcription factor ICER (inducible cAMP early repressor). In the present study, we examined CD4(+)CD25(-) Teff cell (effector T-cell) and CD4(+)CD25(+) Treg cell immune responses in Crem (cAMP-response-element modulator) gene-deficient mice which lack ICER (Crem(-/-)/ICER-deficient mice). ICER deficiency did not significantly alter the frequency or number of Treg cells and Teff cells. Treg cells or a pharmacological increase in cAMP suppressed Teff cells from Crem(+/+) and Crem(-/-)/ICER-deficient mice to an equivalent degree, demonstrating that ICER is dispensable in these functions. Additionally, activating the cAMP effector Epac (exchange protein directly activated by cAMP) suppressed Teff cells. Treg cells expressed low levels of all cyclic nucleotide Pde (phosphodiesterase) genes tested, but high levels of Epac. These data identify ICER as a redundant mediator of Treg cells and cAMP action on Teff cells and suggest that Epac may function as an alternative effector to promote cAMP-dependent Teff cell suppression.


Subject(s)
Cyclic AMP Response Element Modulator/immunology , Cyclic AMP-Dependent Protein Kinases/immunology , Cyclic AMP/immunology , Guanine Nucleotide Exchange Factors/immunology , Immune Tolerance/physiology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Proliferation/physiology , Cyclic AMP/genetics , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Female , Guanine Nucleotide Exchange Factors/genetics , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/cytology
14.
Cell Signal ; 25(9): 1804-12, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23707530

ABSTRACT

Antiviral responses can be triggered by the cytoplasmic RNA helicase RIG-I that binds to viral RNA. RIG-I-mediated signaling stimulates the transcription factors IRF3 and NF-κB and their activation mechanisms have been intensively studied. Here we examined Sendai virus (SV)-mediated activation of the transcription factor CREB and the role of its feedback repressor ICER in production of endogenous antiviral proteins. We show that SV infection and the mitochondrial adapter protein MAVS promote CREB phosphorylation that is dependent upon p38 MAPK and MK2. ICER is induced by CREB and acts as a feedback repressor of CRE-dependent transcription. We found that SV infection stimulated induction of ICER mRNA and protein expression. Surprisingly, ectopic expression and siRNA-mediated knockdown of ICER revealed that ICER is a positive regulator of the production of antiviral IFN-ß and IP10 during SV infection. In contrast, ICER did not affect SV-elicited phosphorylation of IRF3, NF-κB or ATF2/c-Jun, transcription factors governing IFN-ß and IP10 synthesis. However, expression of ICER increased total IRF3 protein levels during SV infection. These results point to a novel role of ICER in antiviral immune signaling acting to increase levels of antiviral effectors.


Subject(s)
Cyclic AMP Response Element Modulator/immunology , DEAD-box RNA Helicases/immunology , Host-Pathogen Interactions , Interferon-beta/immunology , Respirovirus Infections/immunology , Sendai virus/physiology , Activating Transcription Factor 2/immunology , Adaptor Proteins, Signal Transducing/immunology , Cyclic AMP/immunology , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element-Binding Protein/immunology , DEAD Box Protein 58 , HEK293 Cells , Humans , Interferon Regulatory Factor-3/immunology , Interferon-beta/genetics , RNA, Messenger/genetics , Receptors, Immunologic , Respirovirus Infections/genetics , Transcriptional Activation , eIF-2 Kinase/immunology , p38 Mitogen-Activated Protein Kinases/immunology
15.
Trends Mol Med ; 19(4): 262-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23491535

ABSTRACT

T lymphocytes from patients with systemic lupus erythematosus (SLE) display a complex array of cellular, molecular, and signaling anomalies, many of which have been attributed to increased expression of the transcriptional regulator cAMP responsive element modulator α (CREMα). Recent evidence indicates that CREMα, in addition to its regulatory functions on gene promoters in T lymphocytes, alters the epigenetic conformation of cytokine genes by interacting with enzymes that control histone methylation and acetylation as well as cytosine-phosphate-guanosine (CpG) DNA methylation. This review summarizes the most recent findings on CREM protein expression in various cell types, in particular its effects on T lymphocyte biology in the context of both health and SLE. We emphasize CREMα as a key molecule that drives autoimmunity.


Subject(s)
Cyclic AMP Response Element Modulator/immunology , Cytokines/genetics , Epigenesis, Genetic , T-Lymphocytes/immunology , Acetylation , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cyclic AMP Response Element Modulator/genetics , Cytokines/biosynthesis , DNA Methylation , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Promoter Regions, Genetic , Signal Transduction , T-Lymphocytes/metabolism
16.
Proc Natl Acad Sci U S A ; 109(41): 16606-11, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-23019580

ABSTRACT

Appropriate expression of IL-2 plays a central role during the priming and differentiation of T cells. A tight balance between IL-2 and the effector cytokine IL-17A is essential for immune homeostasis. Epigenetic mechanisms have been documented as a key component of cytokine regulation during lineage commitment. The molecular mechanisms that induce chromatin remodeling are less well understood. We investigated epigenetic regulators that mediate the diametric expression of IL-2 and IL-17A in naive, central memory, and effector memory CD4(+) T cells. We demonstrate that cAMP response modulator (CREM)α contributes to epigenetic remodeling of IL2 in effector memory T cells through the recruitment of DNMT3a. CREMα also reduces CpG-DNA methylation of the IL17A promoter. CREMα expression is regulated at the epigenetic level by CpG-DNA methylation, which allows increased CREMα expression in effector memory CD4(+) T cells. T cells from patients with systemic lupus erythematosus (SLE) express increased levels of CREMα and exhibit a phenotype that is similar to effector memory CD4(+) T cells with epigenetically predetermined expression patterns of IL-2 and IL-17A. We conclude that CREMα mediates epigenetic remodeling of the IL2 and IL17A gene during T-cell differentiation in favor of effector memory T cells in health and disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cyclic AMP Response Element Modulator/immunology , Interleukin-17/immunology , Interleukin-2/immunology , Lupus Erythematosus, Systemic/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Lineage/immunology , Cells, Cultured , CpG Islands/genetics , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/immunology , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Methyltransferase 3A , Flow Cytometry , Gene Expression , HEK293 Cells , Humans , Immunologic Memory/immunology , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-2/genetics , Interleukin-2/metabolism , Jurkat Cells , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
17.
Cell Immunol ; 276(1-2): 10-5, 2012.
Article in English | MEDLINE | ID: mdl-22560675

ABSTRACT

Systemic lupus erythematosus (SLE) is a complex autoimmune disease. Immune complex, autoantibodies and autoreactive lymphocytes are involved in manifestations of SLE. Recently, investigations have indicated that expression of the transcription factor cAMP responsive element modulator (CREM) is abnormal in T cells and might play an important role in the pathogenesis of SLE. CREM has much influence on the promoters, such as IL-2, c-fos, TCR ζ, and SYK. Moreover, activity of CREM itself has been demonstrated, particularly with an auto-regulatory feedback mechanism. Therefore, we will discuss the association of CREM and SLE based on current knowledge to unravel the mechanism of CREM performance.


Subject(s)
Cyclic AMP Response Element Modulator/immunology , Lupus Erythematosus, Systemic/immunology , Animals , Cyclic AMP/immunology , Cyclic AMP/metabolism , Cyclic AMP Response Element Modulator/metabolism , Humans , Interleukins/immunology , Lupus Erythematosus, Systemic/metabolism , Promoter Regions, Genetic , T-Lymphocytes/immunology
18.
J Immunol ; 188(12): 5833-7, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22586041

ABSTRACT

Although global MyD88 deficiency attenuates lethal inflammation in sepsis, cell-specific functions of MyD88 remain largely unknown. Using mice with selective expression of MyD88 in myeloid cells (Myd88(MYEL)), we show that, during polymicrobial septic peritonitis, both myeloid and nonmyeloid cells contribute to systemic inflammation, whereas myeloid cell MyD88 was sufficient to fully establish the peritoneal cytokine response. Importantly, Myd88(MYEL) mice developed markedly aggravated liver injury that was linked to impaired upregulation of cellular inhibitor of apoptosis protein 2 and an excessive production of TNF-α. Upregulation of inducible cAMP early repressor (ICER), a known transcriptional repressor of the Tnfa gene, was impaired in Myd88(MYEL) mice. Moreover, Myd88(MYEL) mice showed enhanced transcription of the Tnfa gene and an excessive production of CCL3, which is also negatively regulated by ICER, but they had normal levels of CXCL1, which is expressed in an ICER-independent manner. Together, these findings suggest a novel protective role for nonmyeloid cell MyD88 in attenuating liver injury during septic peritonitis.


Subject(s)
Myeloid Differentiation Factor 88/immunology , Peritonitis/immunology , Sepsis/immunology , Animals , Cyclic AMP Response Element Modulator/immunology , Enzyme-Linked Immunosorbent Assay , Female , Inflammation/immunology , Inflammation/metabolism , Inhibitor of Apoptosis Proteins/immunology , Inhibitor of Apoptosis Proteins/metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Myeloid Cells/immunology , Myeloid Cells/metabolism , Myeloid Differentiation Factor 88/metabolism , Peritonitis/metabolism , Peritonitis/pathology , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/metabolism , Sepsis/pathology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
19.
Eur J Immunol ; 42(4): 912-23, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22531917

ABSTRACT

Gastrointestinal inflammation is mediated by the pro-inflammatory mediators interleukin-8 (IL-8) and prostaglandin E(2) (PGE(2) ). PGE(2) binding and coupling through EP2/4 receptor subtypes on colonic epithelial cells stimulates cyclic adenosine monophosphate (cAMP) and IL-8 production. Here we determined the mechanisms whereby PGE(2) regu-lates IL-8 in Caco2 colonic epithelial cells and in cells over-expressing the EP2/4 receptors (EP2S/EP4S). PGE(2) coupling through EP2 activated the transcription factor inducible cAMP early repressor (ICER), whereas coupling through EP4 receptors activated the cyclic AMP-responsive element-binding protein (CREB). Activation of CREB in Caco2/EP2S was protein kinase A (PKA) dependent, whereas in EP4S cells, activation of CREB occurred through the PKA and phosphatidylinositol 3-kinase pathways. Since ICER lacks the transactivation domain, it functions as a transcription repressor as opposed to CREB. PGE(2) coupling through EP2/4 receptors can therefore acts in an opposing manner to either decrease (EP2) or promote IL-8 expression by recruiting CREB-binding protein (CBP) (EP4), which formed a multiprotein IL-8 enhanceosome. A novel half CRE (167CRE) and a composite NFAT1-AP1-like site in the IL-8 promoter participated in binding and complex formation as confirmed by mutagenesis and expression studies. These data unravel the mechanisms by which expression of IL-8 is controlled by different signalling pathways that are activated by PGE(2) but acting through different EP receptors.


Subject(s)
Colon/immunology , Dinoprostone/immunology , Epithelial Cells/immunology , Gene Expression Regulation/immunology , Interleukin-8/immunology , Intestinal Mucosa/immunology , Caco-2 Cells , Colon/cytology , Colon/metabolism , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/immunology , Cyclic AMP Response Element Modulator/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/immunology , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/immunology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/genetics , Dinoprostone/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation/genetics , Humans , Interleukin-8/biosynthesis , Interleukin-8/genetics , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/immunology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Response Elements/genetics , Response Elements/immunology , Signal Transduction/genetics , Signal Transduction/immunology
20.
J Biol Chem ; 287(7): 4715-25, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22184122

ABSTRACT

The proinflammatory cytokines IL-17A and IL-17F are primarily produced by Th17 lymphocytes. Both are involved in host defense mechanisms against bacterial and fungal pathogens and contribute to the development of various autoimmune diseases. T lymphocytes from patients with systemic lupus erythematosus (SLE) display increased expression of transcription factor cAMP-responsive element modulator α (CREMα), which has been documented to account for aberrant T cell function and contributes to the pathogenesis of SLE. Here, we provide evidence that IL-17F expression is reduced in SLE T cells. We demonstrate that CREMα binds to a yet unidentified CRE site within the proximal promoter. This results in reduced IL-17F expression in SLE T lymphocytes and is independent of activating epigenetic patterns (increased histone H3 Lys-18 acetylation, reduced histone H3 Lys-27 trimethylation, and CpG-DNA demethylation). Forced CREMα expression in human T lymphocytes results in reduced IL-17F expression. Our findings demonstrate extended involvement of CREMα in cytokine dysregulation in SLE by contributing to a disrupted balance between IL-17A and IL-17F. An increased IL-17A/IL-17F ratio may aggravate the proinflammatory phenotype of SLE.


Subject(s)
Cyclic AMP Response Element Modulator/metabolism , Epigenesis, Genetic , Interleukin-17/biosynthesis , Lupus Erythematosus, Systemic/metabolism , Response Elements , Th17 Cells/metabolism , Acetylation , Adult , CpG Islands/immunology , Cyclic AMP Response Element Modulator/immunology , Female , Histones/immunology , Histones/metabolism , Humans , Interleukin-17/immunology , Jurkat Cells , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Methylation , Middle Aged , Th17 Cells/immunology , Th17 Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...