Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Molecules ; 26(13)2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34203519

ABSTRACT

Histone acetylation is an epigenetic mechanism that regulates the expression of various genes, such as natural killer group 2, member D (NKG2D) ligands. These NKG2D ligands are the key molecules that activate immune cells expressing the NKG2D receptor. It has been observed that cancer cells overexpress histone deacetylases (HDACs) and show reduced acetylation of nuclear histones. Furthermore, HDAC inhibitors are known to upregulate the expression of NKG2D ligands. Humans have 18 known HDAC enzymes that are divided into four classes. At present, it is not clear which types of HDAC are involved in the expression of NKG2D ligands. We hypothesized that specific types of HDAC genes might be responsible for altering the expression of NKG2D ligands. In this study, we monitored the expression of NKG2D ligands and major histocompatibility complex (MHC) class I molecules in lung cancer cells which were treated with six selective HDAC inhibitors and specific small interfering RNAs (siRNAs). We observed that treatment with FK228, which is a selective HDAC1/2 inhibitor, also known as Romidepsin, induced NKG2D ligand expression at the transcriptional and proteomic levels in two different lung cancer cell lines. It also caused an increase in the susceptibility of NCI-H23 cells to NK cells. Silencing HDAC1 or HDAC2 using specific siRNAs increased NKG2D ligand expression. In conclusion, it appears that HDAC1 and HDAC2 might be the key molecules regulating the expression of NKG2D ligands. These results imply that specifically inhibiting HDAC1 and HDAC2 could induce the expression of NKG2D ligands and improve the NK cell-mediated anti-cancer immunity.


Subject(s)
Gene Expression Regulation, Neoplastic/immunology , Histone Deacetylase 1/immunology , Histone Deacetylase 2/immunology , Immunity, Cellular/immunology , Lung Neoplasms/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasm Proteins/immunology , A549 Cells , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Humans , Killer Cells, Natural , Lung Neoplasms/genetics , Lung Neoplasms/pathology , NK Cell Lectin-Like Receptor Subfamily K/genetics , Neoplasm Proteins/genetics
2.
J Clin Invest ; 130(7): 3717-3733, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32191636

ABSTRACT

T follicular helper (Tfh) cells are indispensable for the formation of germinal center (GC) reactions, whereas T follicular regulatory (Tfr) cells inhibit Tfh-mediated GC responses. Aberrant activation of Tfh cells contributes substantially to the pathogenesis of autoimmune diseases, such as systemic lupus erythematosus (SLE). Nonetheless, the molecular mechanisms mitigating excessive Tfh cell differentiation are not fully understood. Herein we demonstrate that the adenovirus E4 promoter-binding protein (E4BP4) mediates a feedback loop and acts as a transcriptional brake to inhibit Tfh cell differentiation. Furthermore, we show that such an immunological mechanism is compromised in patients with SLE. Establishing mice with either conditional knockout (cKO) or knockin (cKI) of the E4bp4 gene in T cells reveals that E4BP4 strongly inhibits Tfh cell differentiation. Mechanistically, E4BP4 regulates Bcl6 transcription by recruiting the repressive epigenetic modifiers HDAC1 and EZH2. E4BP4 phosphorylation site mutants have limited capability with regard to inhibiting Tfh cell differentiation. In SLE, we detected impaired phosphorylation of E4BP4, finding that this compromised transcription factor is positively correlated with disease activity. These findings unveiled molecular mechanisms by which E4BP4 restrains Tfh cell differentiation, whose compromised function is associated with uncontrolled autoimmune reactions in SLE.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Cell Differentiation/immunology , Lupus Erythematosus, Systemic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/immunology , Female , Histone Deacetylase 1/genetics , Histone Deacetylase 1/immunology , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/immunology , T-Lymphocytes, Helper-Inducer/pathology
3.
J Immunol ; 204(5): 1201-1213, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31932499

ABSTRACT

Vitamin D can modulate the innate and adaptive immune system. Vitamin D deficiency has been associated with various autoimmune diseases. Th9 cells are implicated in the pathogenesis of numerous autoimmune diseases. Thus, we investigated the role of calcitriol (active metabolite of vitamin D) in the regulation of Th9 cell differentiation. In this study, we have unraveled the molecular mechanisms of calcitriol-mediated regulation of Th9 cell differentiation. Calcitriol significantly diminished IL-9 secretion from murine Th9 cells associated with downregulated expression of the Th9-associated transcription factor, PU.1. Ectopic expression of VDR in Th9 cells attenuated the percentage of IL-9-secreting cells. VDR associated with PU.1 in Th9 cells. Using a series of mutations, we were able to dissect the VDR domain involved in the regulation of the Il9 gene. The VDR-PU.1 interaction prevented the accessibility of PU.1 to the Il9 gene promoter, thereby restricting its expression. However, the expression of Foxp3, regulatory T cell-specific transcription factor, was enhanced in the presence of calcitriol in Th9 cells. When Th9 cells are treated with both calcitriol and trichostatin A (histone deacetylase inhibitor), the level of IL-9 reached to the level of wild-type untreated Th9 cells. Calcitriol attenuated specific histone acetylation at the Il9 gene. In contrast, calcitriol enhanced the recruitment of the histone modifier HDAC1 at the Il9 gene promoter. In summary, we have identified that calcitriol blocked the access of PU.1 to the Il9 gene by reducing its expression and associating with it as well as regulated the chromatin of the Il9 gene to regulate expression.


Subject(s)
Calcitriol/pharmacology , Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Histone Deacetylase 1/immunology , Interleukin-9/immunology , Proto-Oncogene Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Trans-Activators/immunology , Acetylation/drug effects , Animals , Cell Differentiation/immunology , Female , Gene Expression Regulation/immunology , Histones/immunology , Mice , Promoter Regions, Genetic/immunology , Receptors, Calcitriol/immunology , T-Lymphocytes, Regulatory/cytology
4.
J Exp Med ; 215(12): 3194-3212, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30463877

ABSTRACT

In contrast to the common role of histone deacetylases (HDACs) for gene repression, HDAC activity provides a required positive function for IFN-stimulated gene (ISG) expression. Here, we show that HDAC1/2 as components of the Sin3A complex are required for ISG transcriptional elongation but not for recruitment of RNA polymerase or transcriptional initiation. Transcriptional arrest by HDAC inhibition coincides with failure to recruit the epigenetic reader Brd4 and elongation factor P-TEFb due to sequestration of Brd4 on hyperacetylated chromatin. Brd4 availability is regulated by an equilibrium cycle between opposed acetyltransferase and deacetylase activities that maintains a steady-state pool of free Brd4 available for recruitment to inducible promoters. An ISG expression signature is a hallmark of interferonopathies and other autoimmune diseases. Combined inhibition of HDAC1/2 and Brd4 resolved the aberrant ISG expression detected in cells derived from patients with two inherited interferonopathies, ISG15 and USP18 deficiencies, defining a novel therapeutic approach to ISG-associated autoimmune diseases.


Subject(s)
Autoimmune Diseases/immunology , Gene Expression Regulation/immunology , Genetic Diseases, Inborn/immunology , Histone Deacetylase 1/immunology , Histone Deacetylase 2/immunology , Nuclear Proteins/immunology , Promoter Regions, Genetic/immunology , Transcription Factors/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Cell Cycle Proteins , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , HEK293 Cells , HeLa Cells , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Humans , Interferons/genetics , Interferons/immunology , Nuclear Proteins/genetics , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/immunology , Transcription Factors/genetics
5.
Nat Commun ; 8: 15818, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28598443

ABSTRACT

Regulatory T (Treg) cells are important for the maintenance of immune homoeostasis and prevention of autoimmune diseases. Epigenetic modifications have been reported to modulate autoimmunity by altering Treg cell fate. Here we show that the H3K4 methyltransferase Ash1l facilitates TGF-ß-induced Treg cell polarization in vitro and protects mice from T cell-mediated colitis in vivo. Ash1l upregulates Smad3 expression by directly targeting Smad3 promoter to increase local H3K4 trimethylation. Furthermore, we identify an lncRNA, namely lnc-Smad3, which interacts with the histone deacetylase HDAC1 and silences Smad3 transcription. After TGF-ß stimulation, activated Smad3 suppresses lnc-Smad3 transcription, thereby recovering the Smad3 promoter accessibility to Ash1l. By revealing the opposite regulatory functions of Ash1l and lnc-Smad3 in Smad3 expression, our data provide insights for the epigenetic control of Treg cell fate to potentially aid in the development of therapeutic intervention for autoimmune diseases.


Subject(s)
Autoimmunity , Cell Polarity , Histone-Lysine N-Methyltransferase/immunology , RNA, Long Noncoding/genetics , Smad3 Protein/genetics , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes/immunology , Adult , Aged , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Epigenesis, Genetic , Gene Expression Regulation , Gene Silencing , Histone Deacetylase 1/genetics , Histone Deacetylase 1/immunology , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/immunology , Humans , Methylation , Mice , Mice, Inbred C57BL , Middle Aged , Promoter Regions, Genetic , RNA, Long Noncoding/immunology , Smad3 Protein/immunology , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Transforming Growth Factor beta/immunology
6.
Blood ; 130(2): 146-155, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28550044

ABSTRACT

Histone acetylation and the families of enzymes responsible for controlling these epigenetic marks have been implicated in regulating T-cell maturation and phenotype. Here, we demonstrate a previously undefined role of histone deacetylase 11 (HDAC11) in regulating T-cell effector functions. Using EGFP-HDAC11 transgenic reporter mice, we found that HDAC11 expression was lower in effector relative to naive and central memory T-cell populations, and activation of resting T cells resulted in its decreased expression. Experiments using HDAC11 knockout (KO) mice revealed that T cells from these mice displayed enhanced proliferation, proinflammatory cytokine production, and effector molecule expression. In addition, HDAC11KO T cells had increased expression of Eomesodermin (Eomes) and TBX21 (Tbet), transcription factors previously shown to regulate inflammatory cytokine and effector molecule production. Conversely, overexpression of HDAC11 resulted in decreased expression of these genes. Chromatin immunoprecipitation showed the presence of HDAC11 at the Eomes and Tbet gene promoters in resting T cells, where it rapidly disassociated following T-cell activation. In vivo, HDAC11KO T cells were refractory to tolerance induction. HDAC11KO T cells also mediated accelerated onset of acute graft-versus-host disease (GVHD) in a murine model, characterized by increased proliferation of T cells and expression of interferon-γ, tumor necrosis factor, and EOMES. In addition, adoptive transfer of HDAC11KO T cells resulted in significantly reduced tumor burden in a murine B-cell lymphoma model. Taken together, these data demonstrate a previously unknown role of HDAC11 as a negative epigenetic regulator of T-cell effector phenotype and function.


Subject(s)
Gene Expression Regulation, Neoplastic , Graft vs Host Disease/immunology , Histone Deacetylase 1/genetics , Lymphoma, B-Cell/immunology , T-Box Domain Proteins/genetics , T-Lymphocytes/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Chromatin/chemistry , Chromatin/metabolism , Chromatin Immunoprecipitation , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Histone Deacetylase 1/deficiency , Histone Deacetylase 1/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Activation , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Mice , Mice, Knockout , Promoter Regions, Genetic , Signal Transduction , T-Box Domain Proteins/immunology , T-Lymphocytes/pathology , T-Lymphocytes/transplantation , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
7.
Cell Mol Immunol ; 13(1): 110-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25683610

ABSTRACT

The disruption of epithelial barrier integrity is an important factor in the pathogenesis of various immune disorders. However, the restitution of the compromised barrier functions is difficult. This study investigates the regulation of TWIK-related potassium channel-1 (Trek1) in the restitution of intestinal epithelial barrier functions. The human colon epithelial cell line T84 was cultured in monolayers and used to observe epithelial barrier functions in vitro. An intestinal allergy mouse model was created. Cytokine levels were determined by enzyme-linked immunosorbent assay and western blotting. The results showed that Trek1 deficiency induced T84 monolayer barrier disruption. Allergic responses markedly suppressed the expression of Trek1 in the intestinal epithelia via activating the mitogen-activated protein kinase pathways and increasing the expression of histone deacetylase-1. The inhibition of histone deacetylase-1 by sodium butyrate or the administration of a butyrate-producing probiotic (Clostridium butyricum) restored the intestinal epithelial barrier functions and markedly enhanced the effect of antigen-specific immunotherapy. The data suggest that Trek1 is required for the maintenance of intestinal epithelial barrier integrity. Allergic responses induce an insufficiency of Trek1 expression in the intestinal epithelia. Trek1 expression facilitates the restoration of intestinal epithelial barrier functions in an allergic environment.


Subject(s)
Butyric Acid/pharmacology , Clostridium butyricum/immunology , Epithelial Cells/immunology , Hypersensitivity/therapy , Intestinal Mucosa/immunology , Potassium Channels, Tandem Pore Domain/immunology , Probiotics/pharmacology , Animals , Cell Line , Cytokines/genetics , Cytokines/immunology , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/pathology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gene Expression Regulation , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Histone Deacetylase 1/immunology , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Hypersensitivity/genetics , Hypersensitivity/immunology , Hypersensitivity/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/immunology , Ovalbumin , Potassium Channels, Tandem Pore Domain/agonists , Potassium Channels, Tandem Pore Domain/genetics , Signal Transduction
8.
Sci Rep ; 5: 17651, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26627845

ABSTRACT

The current therapy on allergic inflammation is unsatisfactory. Probiotics improve the immunity in the body. This study aims to test a hypothesis that administration with Clostridium butyricum (C. butyricum) enforces the effect of specific immunotherapy (SIT) on intestinal allergic inflammation. In this study, an ovalbumin (OVA) specific allergic inflammation mouse model was created. The mice were treated with SIT or/and C. butyricum. The results showed that the intestinal allergic inflammation was only moderately alleviated by SIT, which was significantly enforced by a combination with C. butyricum; treating with C. butyricum alone did not show much inhibitory efficacy. The increase in the frequency of the interleukin (IL)-10-producing OVA-specific B cell (OVAsBC) was observed in mice in parallel to the inhibitory effect on the intestinal allergic inflammation. The in vitro treatment of the OVAsBCs with OVA increased the histone deacetylase-1 (HDAC1) phosphorylation, modulated the transcription of the Bcl6 gene, and triggered the OVAsBCs to differentiate to the IgE-producing plasma cells. Exposure to both OVA and butyrate sodium in the culture increased the expression of IL-10 in OVAsBCs. In conclusion, administration with C. butyricum enforces the inhibitory effect of SIT on allergic inflammation in the mouse intestine.


Subject(s)
Clostridium butyricum , Hypersensitivity/therapy , Immunotherapy , Intestines/immunology , Probiotics/pharmacokinetics , Animals , Histone Deacetylase 1/genetics , Histone Deacetylase 1/immunology , Hypersensitivity/genetics , Hypersensitivity/immunology , Hypersensitivity/pathology , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Interleukin-10/genetics , Interleukin-10/immunology , Intestines/pathology , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Plasma Cells/immunology , Plasma Cells/pathology
9.
Immunobiology ; 220(10): 1136-40, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26074062

ABSTRACT

BACKGROUND AND AIMS: The prevalence of lung cancer is increasing in the recent decades. The underlying mechanism is unclear. The insulin-like growth factor (IGF) and p53 protein are important molecules involving the tumor immunity. This study aims to investigate the role of IGF intervene the radiation-induced lung cancer apoptosis. METHODS: Lung cancer cells were isolated from surgically removed lung cancer tissue. The lung cancer cell lines, A549 cells and H23 cells were irradiated. The expression of IGF1 receptor (IGF1R) by the lung cancer cells, and apoptosis, were assessed by flow cytometry. RESULTS: The results showed that human lung cancer cells expressed IGF1R. IGF1R played a critical role in the radiation-induced lung cancer cell apoptosis. The histone deacetylase-1 (HDAC1) phosphorylation was up regulated by irradiation. The phosphorylated HDAC1 bound the p53 promoter to inhibit the gene transcription, which was abolished by the presence of an inhibitor of HDAC1 or a STAT3 inhibitor. CONCLUSION: The data suggest that activation of IGF1R plays a critical role in the radioresistance, which can be prevented in the presence of the inhibitors of HDAC1 or STAT3 inhibitors.


Subject(s)
Apoptosis/immunology , Gamma Rays , Gene Expression Regulation, Neoplastic/radiation effects , Lung Neoplasms/immunology , Neoplasm Proteins/immunology , Radiation Tolerance/immunology , Receptors, Somatomedin/immunology , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/immunology , Histone Deacetylase Inhibitors/pharmacology , Humans , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Neoplasm Proteins/antagonists & inhibitors , Radiation Tolerance/drug effects , Receptor, IGF Type 1 , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/immunology
10.
Int Immunopharmacol ; 28(2): 813-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25864621

ABSTRACT

CD4(+) helper T cells and CD8(+) cytotoxic T cells form the two major subsets of peripheral T lymphocytes. Helper T cells fulfill crucial roles in the activation and coordination of the immune response, while cytotoxic T cells kill virus-infected or tumor cells. Recent data suggest that the lineage identify of helper T cells is not fixed and that CD4(+) T cells under certain physiological conditions can be reprogrammed to express CD8 lineage genes and to develop into intestinal intraepithelial CD4(+) cytotoxic T lymphocytes that lack the expression of the key helper T cell lineage commitment factor ThPOK. Moreover, the analysis of mice with a conditional deletion of the transcription factor ThPOK or the histone deacetylases HDAC1 and HDAC2 indicated that CD8 lineage genes are actively repressed in CD4(+) T cells in order to maintain the lineage integrity of helper T cells. In this review I summarize recent studies that indicate plasticity of CD4(+) T cells towards a CTL program and that demonstrate that ThPOK and HDAC1-HDAC2 are part of a transcriptional regulatory circuit that counteracts the activity of the transcription factor Runx3 to maintain CD4(+) T cell lineage integrity.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Cell Plasticity , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Lineage , Core Binding Factor alpha Subunits/immunology , DNA-Binding Proteins/immunology , Histone Deacetylase 1/immunology , Histone Deacetylase 2/immunology , Humans , Intestines/immunology , Transcription Factors/immunology
11.
PLoS One ; 9(3): e91083, 2014.
Article in English | MEDLINE | ID: mdl-24595367

ABSTRACT

Bronchopulmonary dysplasia (BPD) is characterized by alveolar simplification with decreased alveolar number and increased airspace. Previous studies suggested that transforming growth factor-α (TGF-α) may contribute to arrested alveolar development in BPD. Histone deacetylases (HDACs) control cellular signaling and gene expression. HDAC2 is crucial for suppression of inflammatory gene expression. Here we investigated whether HDAC2 was involved in the arrest of alveolarization, as well as the ability of HDAC2 to regulate TGF-α expression in a rat model of BPD induced by intra-amniotic injection of lipopolysaccharide (LPS). Results showed that LPS exposure led to a suppression of both HDAC1 and HDAC2 expression and activity, induced TGF-α expression, and disrupted alveolar morphology. Mechanistic studies showed that overexpression of HDAC2, but not HDAC1, suppressed LPS-induced TGF-α expression. Moreover, the HDAC inhibitor TSA or downregulation of HDAC2 by siRNA both significantly increased TGF-α expression in cultured myofibroblasts. Finally, preservation of HDAC activity by theophylline treatment improved alveolar development and attenuated TGF-α release. Together, these findings indicate that attenuation of TGF-α-mediated effects in the lung by enhancing HDAC2 may have a therapeutic effect on treating BPD.


Subject(s)
Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/immunology , Histone Deacetylase 2/genetics , Lipopolysaccharides/immunology , Lung/pathology , Transforming Growth Factor alpha/genetics , Up-Regulation , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/pathology , Cell Line , Down-Regulation , Female , Histone Deacetylase 1/genetics , Histone Deacetylase 1/immunology , Histone Deacetylase 2/immunology , Humans , Lipopolysaccharides/administration & dosage , Lung/immunology , Lung/metabolism , Rats , Transforming Growth Factor alpha/immunology
12.
Nat Neurosci ; 16(8): 1008-15, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23852118

ABSTRACT

Defects in DNA repair have been linked to cognitive decline with age and neurodegenerative disease, yet the mechanisms that protect neurons from genotoxic stress remain largely obscure. We sought to characterize the roles of the NAD(+)-dependent deacetylase SIRT1 in the neuronal response to DNA double-strand breaks (DSBs). We found that SIRT1 was rapidly recruited to DSBs in postmitotic neurons, where it showed a synergistic relationship with ataxia telangiectasia mutated (ATM). SIRT1 recruitment to breaks was ATM dependent; however, SIRT1 also stimulated ATM autophosphorylation and activity and stabilized ATM at DSB sites. After DSB induction, SIRT1 also bound the neuroprotective class I histone deacetylase HDAC1. We found that SIRT1 deacetylated HDAC1 and stimulated its enzymatic activity, which was necessary for DSB repair through the nonhomologous end-joining pathway. HDAC1 mutations that mimic a constitutively acetylated state rendered neurons more susceptible to DNA damage, whereas pharmacological SIRT1 activators that promoted HDAC1 deacetylation also reduced DNA damage in two mouse models of neurodegeneration. We propose that SIRT1 is an apical transducer of the DSB response and that SIRT1 activation offers an important therapeutic avenue in neurodegeneration.


Subject(s)
Cell Cycle Proteins/physiology , DNA Breaks, Double-Stranded , DNA End-Joining Repair/physiology , DNA-Binding Proteins/physiology , Genomic Instability , Histone Deacetylase 1/physiology , Neurons/metabolism , Protein Serine-Threonine Kinases/physiology , Sirtuin 1/physiology , Tumor Suppressor Proteins/physiology , Acetylation , Amino Acid Sequence , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Line , Cerebral Cortex/cytology , Comet Assay , Enzyme Activation/drug effects , Etoposide/pharmacology , Genetic Vectors , HEK293 Cells , Hippocampus/cytology , Histone Deacetylase 1/genetics , Histone Deacetylase 1/immunology , Humans , Mice , Mice, Transgenic , Molecular Sequence Data , Phosphorylation , Protein Interaction Mapping , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/physiology , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/genetics
13.
Mucosal Immunol ; 4(4): 409-19, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21248725

ABSTRACT

Gingival epithelia utilize multiple signaling pathways to regulate innate immune responses to various oral bacteria, but little is understood about how these bacteria alter epithelial epigenetic status. In this study we report that DNA methyltransferase (DNMT1) and histone deacetylase expression were decreased in gingival epithelial cells treated with oral pathogen Porphyromonas gingivalis and nonpathogen Fusobacterium nucleatum. Pretreatment with trichostatin A and sodium butyrate, which increase acetylation of chromatin histones, significantly enhanced the gene expression of antimicrobial proteins human ß-defensin 2 (hBD2) and CC chemokine ligand 20 (CCL20) in response to both bacterial challenges. Pretreatment with DNMT inhibitor 5'-azacytidine increased hBD2 and CCL20 expression in response to F. nucleatum, but not to P. gingivalis. Furthermore, we observed a differential pattern of protein levels of H3K4me3, which has been associated with chromatin remodeling and activation of gene transcription, in response to P. gingivalis vs. F. nucleatum. This study provides a new insight into the bacteria-specific innate immune responses via epigenetic regulation.


Subject(s)
Bacteroidaceae Infections/immunology , Chemokine CCL20/metabolism , Epigenomics , Epithelial Cells , Gene Expression Regulation , Gingiva/immunology , beta-Defensins/metabolism , Chemokine CCL20/genetics , Chromatin/metabolism , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/immunology , DNA Methylation/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Fusobacterium nucleatum/immunology , Gene Expression Regulation/immunology , Gingiva/cytology , Gingiva/microbiology , Histone Deacetylase 1/genetics , Histone Deacetylase 1/immunology , Histone Deacetylase 2/genetics , Histone Deacetylase 2/immunology , Histones/metabolism , Humans , Immunity, Innate/immunology , Inflammation/genetics , Inflammation/immunology , Interleukin-8/metabolism , Methylation , Porphyromonas gingivalis/immunology , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , beta-Defensins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL