Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Mol Cell ; 83(19): 3421-3437.e11, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37751740

ABSTRACT

The nuclear receptor co-repressor (NCoR) complex mediates transcriptional repression dependent on histone deacetylation by histone deacetylase 3 (HDAC3) as a component of the complex. Unexpectedly, we found that signaling by the receptor activator of nuclear factor κB (RANK) converts the NCoR/HDAC3 co-repressor complex to a co-activator of AP-1 and NF-κB target genes that are required for mouse osteoclast differentiation. Accordingly, the dominant function of NCoR/HDAC3 complexes in response to RANK signaling is to activate, rather than repress, gene expression. Mechanistically, RANK signaling promotes RNA-dependent interaction of the transcriptional co-activator PGC1ß with the NCoR/HDAC3 complex, resulting in the activation of PGC1ß and inhibition of HDAC3 activity for acetylated histone H3. Non-coding RNAs Dancr and Rnu12, which are associated with altered human bone homeostasis, promote NCoR/HDAC3 complex assembly and are necessary for RANKL-induced osteoclast differentiation in vitro. These findings may be prototypic for signal-dependent functions of NCoR in other biological contexts.


Subject(s)
Osteoclasts , RNA , Humans , Mice , Animals , Co-Repressor Proteins/genetics , Osteoclasts/metabolism , RANK Ligand/genetics , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Gene Expression
2.
Proc Natl Acad Sci U S A ; 121(2): e2316104121, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38165941

ABSTRACT

The nuclear receptor corepressor (NCoR) forms a complex with histone deacetylase 3 (HDAC3) that mediates repressive functions of unliganded nuclear receptors and other transcriptional repressors by deacetylation of histone substrates. Recent studies provide evidence that NCoR/HDAC3 complexes can also exert coactivator functions in brown adipocytes by deacetylating and activating PPARγ coactivator 1α (PGC1α) and that signaling via receptor activator of nuclear factor kappa-B (RANK) promotes the formation of a stable NCoR/HDAC3/PGC1ß complex that coactivates nuclear factor kappa-B (NFκB)- and activator protein 1 (AP-1)-dependent genes required for osteoclast differentiation. Here, we demonstrate that activation of Toll-like receptor (TLR) 4, but not TLR3, the interleukin 4 (IL4) receptor nor the Type I interferon receptor, also promotes assembly of an NCoR/HDAC3/PGC1ß coactivator complex. Receptor-specific utilization of TNF receptor-associated factor 6 (TRAF6) and downstream activation of extracellular signal-regulated kinase 1 (ERK1) and TANK-binding kinase 1 (TBK1) accounts for the common ability of RANK and TLR4 to drive assembly of an NCoR/HDAC3/PGC1ß complex in macrophages. ERK1, the p65 component of NFκB, and the p300 histone acetyltransferase (HAT) are also components of the induced complex and are associated with local histone acetylation and transcriptional activation of TLR4-dependent enhancers and promoters. These observations identify a TLR4/TRAF6-dependent signaling pathway that converts NCoR from a corepressor of nuclear receptors to a coactivator of NFκB and AP-1 that may be relevant to functions of NCoR in other developmental and homeostatic processes.


Subject(s)
Histones , TNF Receptor-Associated Factor 6 , Transcriptional Activation , Co-Repressor Proteins/genetics , Histones/genetics , Histones/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor AP-1/metabolism , Toll-Like Receptor 4/metabolism , Signal Transduction , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism
3.
Nature ; 514(7521): 257-61, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25119036

ABSTRACT

Homeodomain proteins, described 30 years ago, exert essential roles in development as regulators of target gene expression; however, the molecular mechanisms underlying transcriptional activity of homeodomain factors remain poorly understood. Here investigation of a developmentally required POU-homeodomain transcription factor, Pit1 (also known as Pou1f1), has revealed that, unexpectedly, binding of Pit1-occupied enhancers to a nuclear matrin-3-rich network/architecture is a key event in effective activation of the Pit1-regulated enhancer/coding gene transcriptional program. Pit1 association with Satb1 (ref. 8) and ß-catenin is required for this tethering event. A naturally occurring, dominant negative, point mutation in human PIT1(R271W), causing combined pituitary hormone deficiency, results in loss of Pit1 association with ß-catenin and Satb1 and therefore the matrin-3-rich network, blocking Pit1-dependent enhancer/coding target gene activation. This defective activation can be rescued by artificial tethering of the mutant R271W Pit1 protein to the matrin-3 network, bypassing the pre-requisite association with ß-catenin and Satb1 otherwise required. The matrin-3 network-tethered R271W Pit1 mutant, but not the untethered protein, restores Pit1-dependent activation of the enhancers and recruitment of co-activators, exemplified by p300, causing both enhancer RNA transcription and target gene activation. These studies have thus revealed an unanticipated homeodomain factor/ß-catenin/Satb1-dependent localization of target gene regulatory enhancer regions to a subnuclear architectural structure that serves as an underlying mechanism by which an enhancer-bound homeodomain factor effectively activates developmental gene transcriptional programs.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism , RNA-Binding Proteins/metabolism , Transcription, Genetic , Animals , Cells, Cultured , Homeodomain Proteins/genetics , Humans , Matrix Attachment Region Binding Proteins/metabolism , Mice , Pituitary Gland/embryology , Pituitary Gland/metabolism , Protein Binding , Transcription Factor Pit-1/genetics , Transcription Factor Pit-1/metabolism , Transcription, Genetic/genetics , beta Catenin/metabolism
4.
Mol Cell ; 46(1): 91-104, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22424771

ABSTRACT

The association between hyperinflammatory states and numerous diseases is widely recognized, but our understanding of the molecular strategies that have evolved to prevent uncontrolled activation of inflammatory responses remains incomplete. Here, we report a critical, nontranscriptional role of GPS2 as a guardian against hyperstimulation of the TNF-α-induced gene program. GPS2 cytoplasmic actions are required to specifically modulate RIP1 ubiquitylation and JNK activation by inhibiting TRAF2/Ubc13 enzymatic activity. In vivo relevance of GPS2 anti-inflammatory role is confirmed by inhibition of TNF-α target genes in macrophages and by improved insulin signaling in the adipose tissue of aP2-GPS2 transgenic mice. As the nontranscriptional role is complemented by GPS2 functioning as positive and negative cofactor for nuclear receptors, in vivo overexpression also results in elevated circulating level of Resistin and development of hepatic steatosis. Together, these studies define GPS2 as a molecular guardian required for precise control of inflammatory responses involved in immunity and homeostasis.


Subject(s)
Adipose Tissue/metabolism , Homeostasis , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Adipose Tissue/immunology , Animals , Cell Line , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/immunology , GTPase-Activating Proteins/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Insulin/genetics , Insulin/immunology , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/immunology , MAP Kinase Kinase 4/metabolism , Macrophages/immunology , Mice , Mice, Transgenic , Resistin/genetics , Resistin/immunology , Resistin/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/immunology , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination/genetics , Ubiquitination/immunology
5.
Proc Natl Acad Sci U S A ; 113(47): 13408-13413, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27810956

ABSTRACT

As a key component of the vertebrate neuroendocrine system, the pituitary gland relies on the progressive and coordinated development of distinct hormone-producing cell types and an invading vascular network. The molecular mechanisms that drive formation of the pituitary vasculature, which is necessary for regulated synthesis and secretion of hormones that maintain homeostasis, metabolism, and endocrine function, remain poorly understood. Here, we report that expression of integrin ß1 in embryonic pituitary epithelial cells is required for angiogenesis in the developing mouse pituitary gland. Deletion of pituitary epithelial integrin ß1 before the onset of angiogenesis resulted in failure of invading endothelial cells to recruit pericytes efficiently, whereas deletion later in embryogenesis led to decreased vascular density and lumen formation. In both cases, lack of epithelial integrin ß1 was associated with a complete absence of vasculature in the pituitary gland at birth. Within pituitary epithelial cells, integrin ß1 directs a large transcriptional program that includes components of the extracellular matrix and associated signaling factors that are linked to the observed non-cell-autonomous effects on angiogenesis. We conclude that epithelial integrin ß1 functions as a critical and canonical regulator of developmental angiogenesis in the pituitary gland, thus providing insight into the long-standing systems biology conundrum of how vascular invasion is coordinated with tissue development.


Subject(s)
Embryonic Development , Epithelial Cells/metabolism , Integrin beta1/metabolism , Neovascularization, Physiologic , Pituitary Gland/cytology , Pituitary Gland/embryology , Animals , Animals, Newborn , Cell Count , Cell Differentiation , Embryonic Development/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Gene Targeting , Integrases/metabolism , Mice , Neovascularization, Physiologic/genetics , Paired Box Transcription Factors/metabolism , Pericytes/cytology , Pericytes/metabolism , Phenotype , Pituitary Gland/metabolism , Sequence Analysis, RNA , Time Factors , Vascular Endothelial Growth Factor C/metabolism
6.
bioRxiv ; 2024 May 22.
Article in English | MEDLINE | ID: mdl-38798402

ABSTRACT

Because most DNA-binding transcription factors (dbTFs), including the architectural regulator CTCF, bind RNA and exhibit di-/multimerization, a central conundrum is whether these distinct properties are regulated post-transcriptionally to modulate transcriptional programs. Here, investigating stress-dependent activation of SIRT1, encoding an evolutionarily-conserved protein deacetylase, we show that induced phosphorylation of CTCF acts as a rheostat to permit CTCF occupancy of low-affinity promoter DNA sites to precisely the levels necessary. This CTCF recruitment to the SIRT1 promoter is eliciting a cardioprotective cardiomyocyte transcriptional activation program and provides resilience against the stress of the beating heart in vivo . Mice harboring a mutation in the conserved low-affinity CTCF promoter binding site exhibit an altered, cardiomyocyte-specific transcriptional program and a systolic heart failure phenotype. This transcriptional role for CTCF reveals that a covalent dbTF modification regulating signal-dependent transcription serves as a previously unsuspected component of the oxidative stress response.

7.
Nature ; 446(7138): 882-7, 2007 Apr 19.
Article in English | MEDLINE | ID: mdl-17392792

ABSTRACT

Precise control of transcriptional programmes underlying metazoan development is modulated by enzymatically active co-regulatory complexes, coupled with epigenetic strategies. One thing that remains unclear is how specific members of histone modification enzyme families, such as histone methyltransferases and demethylases, are used in vivo to simultaneously orchestrate distinct developmental gene activation and repression programmes. Here, we report that the histone lysine demethylase, LSD1--a component of the CoREST-CtBP co-repressor complex--is required for late cell-lineage determination and differentiation during pituitary organogenesis. LSD1 seems to act primarily on target gene activation programmes, as well as in gene repression programmes, on the basis of recruitment of distinct LSD1-containing co-activator or co-repressor complexes. LSD1-dependent gene repression programmes can be extended late in development with the induced expression of ZEB1, a Krüppel-like repressor that can act as a molecular beacon for recruitment of the LSD1-containing CoREST-CtBP co-repressor complex, causing repression of an additional cohort of genes, such as Gh, which previously required LSD1 for activation. These findings suggest that temporal patterns of expression of specific components of LSD1 complexes modulate gene regulatory programmes in many mammalian organs.


Subject(s)
Down-Regulation/genetics , Gene Expression Regulation, Developmental , Oxidoreductases, N-Demethylating/metabolism , Animals , Cell Differentiation , Growth Hormone/genetics , Histone Demethylases , Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Lactotrophs/metabolism , Mice , Oxidoreductases, N-Demethylating/deficiency , Oxidoreductases, N-Demethylating/genetics , Pituitary Gland/cytology , Pituitary Gland/metabolism , Transcriptional Activation , Zinc Finger E-box-Binding Homeobox 1
8.
Nat Struct Mol Biol ; 30(2): 148-158, 2023 02.
Article in English | MEDLINE | ID: mdl-36747093

ABSTRACT

Enhancer activation serves as the main mechanism regulating signal-dependent transcriptional programs, ensuring cellular plasticity, yet central questions persist regarding their mechanism of activation. Here, by successfully mapping topoisomerase I-DNA covalent complexes genome-wide, we find that most, if not all, acutely activated enhancers, including those induced by 17ß-estradiol, dihydrotestosterone, tumor necrosis factor alpha and neuronal depolarization, are hotspots for topoisomerase I-DNA covalent complexes, functioning as epigenomic signatures read by the classic DNA damage sensor protein, Ku70. Ku70 in turn nucleates a heterochromatin protein 1 gamma (HP1γ)-mediator subunit Med26 complex to facilitate acute, but not chronic, transcriptional activation programs. Together, our data uncover a broad, unappreciated transcriptional code, required for most, if not all, acute signal-dependent enhancer activation events in both mitotic and postmitotic cells.


Subject(s)
DNA Topoisomerases, Type I , Enhancer Elements, Genetic , DNA , DNA Topoisomerases, Type I/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Ku Autoantigen/metabolism
9.
Stem Cell Reports ; 5(6): 1196-1209, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26651607

ABSTRACT

Although SOX2(+) stem cells are present in the postnatal pituitary gland, how they are regulated molecularly and whether they are required for pituitary functions remain unresolved questions. Using a conditional knockout animal model, here we demonstrate that ablation of the canonical Notch signaling in the embryonic pituitary gland leads to progressive depletion of the SOX2(+) stem cells and hypoplastic gland. Furthermore, we show that the SOX2(+) stem cells initially play a significant role in contributing to postnatal pituitary gland expansion by self-renewal and differentiating into distinct lineages in the immediate postnatal period. However, we found that within several weeks postpartum, the SOX2(+) stem cells switch to an essentially dormant state and are no longer required for homeostasis/tissue adaptation. Our results present a dynamic tissue homeostatic model in which stem cells provide an initial contribution to the growth of the neonatal pituitary gland, whereas the mature gland can be maintained in a stem cell-independent fashion.


Subject(s)
Pituitary Gland/cytology , Receptors, Notch/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction , Stem Cells/cytology , Animals , Cell Differentiation , Cell Proliferation , Female , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice, Transgenic , Mutation , Pituitary Gland/metabolism , SOXB1 Transcription Factors/genetics , Stem Cells/metabolism
10.
Neuron ; 86(3): 696-710, 2015 May 06.
Article in English | MEDLINE | ID: mdl-25892301

ABSTRACT

One of the exceptional properties of the brain is its ability to acquire new knowledge through learning and to store that information through memory. The epigenetic mechanisms linking changes in neuronal transcriptional programs to behavioral plasticity remain largely unknown. Here, we identify the epigenetic signature of the neuronal enhancers required for transcriptional regulation of synaptic plasticity genes during memory formation, linking this to Reelin signaling. The binding of Reelin to its receptor, LRP8, triggers activation of this cohort of LRP8-Reelin-regulated neuronal (LRN) enhancers that serve as the ultimate convergence point of a novel synapse-to-nucleus pathway. Reelin simultaneously regulates NMDA-receptor transmission, which reciprocally permits the required γ-secretase-dependent cleavage of LRP8, revealing an unprecedented role for its intracellular domain in the regulation of synaptically generated signals. These results uncover an in vivo enhancer code serving as a critical molecular component of cognition and relevant to psychiatric disorders linked to defects in Reelin signaling.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Conditioning, Classical/physiology , Extracellular Matrix Proteins/metabolism , LDL-Receptor Related Proteins/metabolism , Memory/physiology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Serine Endopeptidases/metabolism , Animals , Bicuculline/pharmacology , CREB-Binding Protein/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Matrix Proteins/genetics , Histone Deacetylases/metabolism , Humans , LDL-Receptor Related Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Molecular , N-Acetylglucosaminyltransferases/genetics , Nerve Tissue Proteins/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Reelin Protein , Serine Endopeptidases/genetics , Signal Transduction/genetics , Synaptic Transmission/drug effects , Synaptic Transmission/genetics
11.
Nat Neurosci ; 18(9): 1256-64, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26214369

ABSTRACT

We found that a neuron-specific isoform of LSD1, LSD1n, which results from an alternative splicing event, acquires a new substrate specificity, targeting histone H4 Lys20 methylation, both in vitro and in vivo. Selective genetic ablation of LSD1n led to deficits in spatial learning and memory, revealing the functional importance of LSD1n in neuronal activity-regulated transcription that is necessary for long-term memory formation. LSD1n occupied neuronal gene enhancers, promoters and transcribed coding regions, and was required for transcription initiation and elongation steps in response to neuronal activity, indicating the crucial role of H4K20 methylation in coordinating gene transcription with neuronal function. Our results indicate that this alternative splicing of LSD1 in neurons, which was associated with altered substrate specificity, serves as a mechanism acquired by neurons to achieve more precise control of gene expression in the complex processes underlying learning and memory.


Subject(s)
Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Memory, Long-Term/physiology , Transcription, Genetic/physiology , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Female , Gene Deletion , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
12.
Proc Natl Acad Sci U S A ; 102(10): 3708-13, 2005 Mar 08.
Article in English | MEDLINE | ID: mdl-15728386

ABSTRACT

The hair-growth cycle, a complex biological system requiring coordinate alterations in gene expression and cellular behavior, provides a challenging model for investigating the interplay of specific transcriptional regulation events. Here we report that the Barx2 homeodomain factor serves as a regulator of hair follicle remodeling (catagen), and loss of Barx2 in mice causes a defect both in the initiation and progression of catagen, resulting in a protracted first catagen, and later, causing short hair in adult gene-deleted mice. Barx2 negatively regulates its own promoter, and our study highlights the role of Barx2 as a repressor in the skin that can, unexpectedly, functionally interact with two WD40-domain factors distantly related to the yeast corepressor Tup1. These two corepressors, transducin-like enhancer of split and transducin beta-like 1, function through distinct and independent interactions with Barx2 for the repression of gene targets, including the Barx2 gene itself, emphasizing the roles of complementary repression strategies in engrailed homology-1 motif-containing homeodomain factors. Together, our data suggest that the hair-remodeling defect of Barx2 mutant mice could be explained, in part, by failure to repress one or more critical target genes.


Subject(s)
Hair Follicle/physiology , Homeodomain Proteins/physiology , Repressor Proteins/physiology , Animals , Cell Differentiation , Gene Expression Profiling , Homeodomain Proteins/genetics , Mice , Skin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL