Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Mol Cell ; 69(3): 398-411.e6, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395062

ABSTRACT

The inflammatory response mediated by nuclear factor κB (NF-κB) signaling is essential for host defense against pathogens. Although the regulatory mechanism of NF-κB signaling has been well studied, the molecular basis for epigenetic regulation of the inflammatory response is poorly understood. Here we identify a new signaling axis of PKCα-LSD1-NF-κB, which is critical for activation and amplification of the inflammatory response. In response to excessive inflammatory stimuli, PKCα translocates to the nucleus and phosphorylates LSD1. LSD1 phosphorylation is required for p65 binding and facilitates p65 demethylation, leading to enhanced stability. In vivo genetic analysis using Lsd1SA/SA mice with ablation of LSD1 phosphorylation and chemical approaches in wild-type mice with inhibition of PKCα or LSD1 activity show attenuated sepsis-induced inflammatory lung injury and mortality. Together, we demonstrate that the PKCα-LSD1-NF-κB signaling cascade is crucial for epigenetic control of the inflammatory response, and targeting this signaling could be a powerful therapeutic strategy for systemic inflammatory diseases, including sepsis.


Subject(s)
Histone Demethylases/metabolism , Protein Kinase C/metabolism , Animals , Cell Nucleus/metabolism , Epigenesis, Genetic/genetics , Histone Demethylases/genetics , Inflammation/metabolism , Methylation , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , Protein Kinase C/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Nature ; 534(7608): 553-7, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27309807

ABSTRACT

Autophagy is a highly conserved self-digestion process, which is essential for maintaining homeostasis and viability in response to nutrient starvation. Although the components of autophagy in the cytoplasm have been well studied, the molecular basis for the transcriptional and epigenetic regulation of autophagy is poorly understood. Here we identify co-activator-associated arginine methyltransferase 1 (CARM1) as a crucial component of autophagy in mammals. Notably, CARM1 stability is regulated by the SKP2-containing SCF (SKP1-cullin1-F-box protein) E3 ubiquitin ligase in the nucleus, but not in the cytoplasm, under nutrient-rich conditions. Furthermore, we show that nutrient starvation results in AMP-activated protein kinase (AMPK)-dependent phosphorylation of FOXO3a in the nucleus, which in turn transcriptionally represses SKP2. This repression leads to increased levels of CARM1 protein and subsequent increases in histone H3 Arg17 dimethylation. Genome-wide analyses reveal that CARM1 exerts transcriptional co-activator function on autophagy-related and lysosomal genes through transcription factor EB (TFEB). Our findings demonstrate that CARM1-dependent histone arginine methylation is a crucial nuclear event in autophagy, and identify a new signalling axis of AMPK-SKP2-CARM1 in the regulation of autophagy induction after nutrient starvation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/genetics , Protein-Arginine N-Methyltransferases/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Signal Transduction , Transcription, Genetic , Animals , Arginine/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line , Cell Nucleus/metabolism , Food Deprivation , Forkhead Box Protein O3 , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Histones/metabolism , Humans , Lysosomes/genetics , Methylation , Mice , Phosphorylation , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/metabolism
3.
Mol Cell ; 39(1): 71-85, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20603076

ABSTRACT

Lysine methylation within histones is crucial for transcriptional regulation and thus links chromatin states to biological outcomes. Although recent studies have extended lysine methylation to nonhistone proteins, underlying molecular mechanisms such as the upstream signaling cascade that induces lysine methylation and downstream target genes modulated by this modification have not been elucidated. Here, we show that Reptin, a chromatin-remodeling factor, is methylated at lysine 67 in hypoxic conditions by the methyltransferase G9a. Methylated Reptin binds to the promoters of a subset of hypoxia-responsive genes and negatively regulates transcription of these genes to modulate cellular responses to hypoxia.


Subject(s)
Carrier Proteins/metabolism , DNA Helicases/metabolism , ATPases Associated with Diverse Cellular Activities , Animals , Cell Hypoxia/genetics , Cell Line , Female , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lysine/metabolism , Methylation , Mice , Models, Biological , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Protein Binding , Xenograft Model Antitumor Assays
4.
Proc Natl Acad Sci U S A ; 108(33): 13510-5, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21825155

ABSTRACT

Pontin is a chromatin remodeling factor that possesses both ATPase and DNA helicase activities. Although Pontin is frequently overexpressed in human cancers of various types and implicated in oncogenic functions, the upstream signaling network leading to the regulation of Pontin that in turn affects transcription of downstream target genes has not been extensively studied. Here, we identify Pontin is methylated by G9a/GLP methyltransferases in hypoxic condition and potentiates HIF-1α-mediated activation by increasing the recruitment of p300 coactivator to a subset of HIF-1α target promoters. Intriguingly, Pontin methylation results in the increased invasive and migratory properties by activating downstream target gene, Ets1. In contrast, inhibition of Pontin methylation results in the suppression of tumorigenic and metastatic properties. Together, our data provide new approaches by targeting Pontin methylation and its downstream targets for the development of therapeutic agents for human cancers.


Subject(s)
Carrier Proteins/metabolism , Chromatin Assembly and Disassembly , DNA Helicases/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , ATPases Associated with Diverse Cellular Activities , Cell Hypoxia , Cell Line, Tumor , Chromatin/genetics , Epigenomics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Methylation , Methyltransferases/metabolism , Neoplasm Proteins/metabolism , Transcription, Genetic
5.
Biochem Biophys Res Commun ; 400(3): 396-402, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20800578

ABSTRACT

B-cell lymphoma 3 (Bcl3) is a proto-oncogene upregulated in a wide range of cancers, including breast cancer. Although Bcl3 is known to promote cell proliferation and inhibit apoptosis, the molecular mechanisms underlying the proto-oncogenic function of Bcl3 have not been completely elucidated. To gain insight into the oncogenic role of Bcl3, we applied a proteomic approach, which led to the identification of C-terminal binding protein 1 (CtBP1) as a binding partner of Bcl3. A PXDLS/R motif embedded in Bcl3 was found to mediate the interaction between Bcl3 and CtBP1, which caused the stabilization of CtBP1 by blocking proteasome-dependent degradation. Apoptotic stimuli-induced degradation of CtBP1 was significantly abolished by the upregulation of Bcl3, leading to the sustained repression of pro-apoptotic gene expression and subsequent inhibition of apoptosis. Intriguingly, a strong positive correlation between the protein levels of Bcl3 and CtBP1 was detected in breast cancer patient samples. Our study reveals a novel combinatorial role for Bcl3 and CtBP1, providing an explanation for the acquisition of resistance to apoptosis in cancer cells, which is a major requirement for cancer development.


Subject(s)
Alcohol Oxidoreductases/metabolism , Apoptosis , Breast Neoplasms/pathology , Cell Proliferation , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Alcohol Oxidoreductases/antagonists & inhibitors , B-Cell Lymphoma 3 Protein , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA-Binding Proteins/antagonists & inhibitors , Enzyme Stability , Female , Humans , Proto-Oncogene Mas , Ubiquitination
6.
Autophagy ; 12(11): 2248-2249, 2016 11.
Article in English | MEDLINE | ID: mdl-27487449

ABSTRACT

Macroautophagy (hereafter referred to as autophagy) is an essential self-digestion process to maintain homeostasis and promote survival in response to starvation. Although the components of autophagy in the cytoplasm have been well studied, little has been known about the fine-tuning mechanism of autophagy through epigenetic regulations. Recently, we identified the histone arginine methyltransferase CARM1 as a new component and followed histone H3R17 dimethylation as a critical epigenetic mark in starvation-induced autophagy. Upon nutrient starvation, CARM1 is stabilized in the nucleus, but not in the cytoplasm, whereas it is constantly degraded under nutrient-rich conditions by the SKP2-containing SCF (SKP1-CUL1-F-box protein) E3 ubiquitin ligase. We further showed that nutrient starvation induces the protein levels and activity of AMPK in the nucleus. Activated AMPK then phosphorylates FOXO3, leading to SKP2 downregulation and increased CARM1 protein levels in the nucleus. Stabilized CARM1 in turn functions as an essential co-activator of TFEB and regulates the expression of autophagy and lysosomal genes. Our findings provide a conceptual advance that activation of specific epigenetic programs is indispensable for a sustained autophagic response, and shed light on a potential therapeutic targeting of the newly identified AMPK-SKP2-CARM1 signaling axis in autophagy-related diseases.


Subject(s)
Autophagy/genetics , Epigenesis, Genetic , Transcription, Genetic , Animals , Humans , Mice , Models, Biological , Protein-Arginine N-Methyltransferases/metabolism , Signal Transduction/genetics
7.
Nat Commun ; 6: 6810, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25857206

ABSTRACT

The actions of transcription factors, chromatin modifiers and noncoding RNAs are crucial for the programming of cell states. Although the importance of various epigenetic machineries for controlling pluripotency of embryonic stem (ES) cells has been previously studied, how chromatin modifiers cooperate with specific transcription factors still remains largely elusive. Here, we find that Pontin chromatin remodelling factor plays an essential role as a coactivator for Oct4 for maintenance of pluripotency in mouse ES cells. Genome-wide analyses reveal that Pontin and Oct4 share a substantial set of target genes involved in ES cell maintenance. Intriguingly, we find that the Oct4-dependent coactivator function of Pontin extends to the transcription of large intergenic noncoding RNAs (lincRNAs) and in particular linc1253, a lineage programme repressing lincRNA, is a Pontin-dependent Oct4 target lincRNA. Together, our findings demonstrate that the Oct4-Pontin module plays critical roles in the regulation of genes involved in ES cell fate determination.


Subject(s)
DNA Helicases/genetics , Epigenesis, Genetic , Mouse Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , RNA, Long Noncoding/genetics , Animals , Cell Differentiation , Chromatin/chemistry , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA Helicases/deficiency , Gene Expression Profiling , Genome-Wide Association Study , Mice , Mice, Knockout , Mouse Embryonic Stem Cells/cytology , Octamer Transcription Factor-3/deficiency , Patched Receptors , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA, Long Noncoding/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tropomyosin/genetics , Tropomyosin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL