Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Haematologica ; 109(6): 1893-1908, 2024 06 01.
Article in English | MEDLINE | ID: mdl-38124661

ABSTRACT

REIIBP is a lysine methyltransferase aberrantly expressed through alternative promoter usage of NSD2 locus in t(4;14)-translocated multiple myeloma (MM). Clinically, t(4;14) translocation is an adverse prognostic factor found in approximately 15% of MM patients. The contribution of REIIBP relative to other NSD2 isoforms as a dependency gene in t(4;14)-translocated MM remains to be evaluated. Here, we demonstrated that despite homology with NSD2, REIIBP displayed distinct substrate specificity by preferentially catalyzing H3K4me3 and H3K27me3, with little activity on H3K36me2. Furthermore, REIIBP was regulated through microRNA by EZH2 in a Dicer-dependent manner, exemplifying a role of REIIBP in SET-mediated H3K27me3. Chromatin immunoprecipitation sequencing revealed chromatin remodeling characterized by changes in genome-wide and loci-specific occupancy of these opposing histone marks, allowing a bidirectional regulation of its target genes. Transcriptomics indicated that REIIBP induced a pro-inflammatory gene signature through upregulation of TLR7, which in turn led to B-cell receptor-independent activation of BTK and driving NFkB-mediated production of cytokines such as IL-6. Activation of this pathway is targetable using Ibrutinib and partially mitigated bortezomib resistance in a REIIBP xenograft model. Mechanistically, REIIBP upregulated TLR7 through eIF3E, and this relied on eIF3E RNA-binding function instead of its canonical protein synthesis activity, as demonstrated by direct binding to the 3'UTR of TLR7 mRNA. Altogether, we provided a rationale that co-existence of different NSD2 isoforms induced diversified oncogenic programs that should be considered in the strategies for t(4;14)-targeted therapy.


Subject(s)
Chromosomes, Human, Pair 14 , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase , Multiple Myeloma , Translocation, Genetic , Humans , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Animals , Mice , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 4/genetics , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Phenotype , Inflammation/genetics , Inflammation/metabolism , Histones/metabolism , Repressor Proteins
2.
Mol Psychiatry ; 26(10): 5766-5788, 2021 10.
Article in English | MEDLINE | ID: mdl-32647257

ABSTRACT

A population of more than six million people worldwide at high risk of Alzheimer's disease (AD) are those with Down Syndrome (DS, caused by trisomy 21 (T21)), 70% of whom develop dementia during lifetime, caused by an extra copy of ß-amyloid-(Aß)-precursor-protein gene. We report AD-like pathology in cerebral organoids grown in vitro from non-invasively sampled strands of hair from 71% of DS donors. The pathology consisted of extracellular diffuse and fibrillar Aß deposits, hyperphosphorylated/pathologically conformed Tau, and premature neuronal loss. Presence/absence of AD-like pathology was donor-specific (reproducible between individual organoids/iPSC lines/experiments). Pathology could be triggered in pathology-negative T21 organoids by CRISPR/Cas9-mediated elimination of the third copy of chromosome 21 gene BACE2, but prevented by combined chemical ß and γ-secretase inhibition. We found that T21 organoids secrete increased proportions of Aß-preventing (Aß1-19) and Aß-degradation products (Aß1-20 and Aß1-34). We show these profiles mirror in cerebrospinal fluid of people with DS. We demonstrate that this protective mechanism is mediated by BACE2-trisomy and cross-inhibited by clinically trialled BACE1 inhibitors. Combined, our data prove the physiological role of BACE2 as a dose-sensitive AD-suppressor gene, potentially explaining the dementia delay in ~30% of people with DS. We also show that DS cerebral organoids could be explored as pre-morbid AD-risk population detector and a system for hypothesis-free drug screens as well as identification of natural suppressor genes for neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Down Syndrome , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Brain/metabolism , Down Syndrome/genetics , Genes, Suppressor , Humans , Organoids/metabolism , Trisomy
3.
PLoS Biol ; 15(2): e1002597, 2017 02.
Article in English | MEDLINE | ID: mdl-28207742

ABSTRACT

Obesity develops when caloric intake exceeds metabolic needs. Promoting energy expenditure represents an attractive approach in the prevention of this fast-spreading epidemic. Here, we report a novel pharmacological strategy in which a natural compound, narciclasine (ncls), attenuates diet-induced obesity (DIO) in mice by promoting energy expenditure. Moreover, ncls promotes fat clearance from peripheral metabolic tissues, improves blood metabolic parameters in DIO mice, and protects these mice from the loss of voluntary physical activity. Further investigation suggested that ncls achieves these beneficial effects by promoting a shift from glycolytic to oxidative muscle fibers in the DIO mice thereby enhancing mitochondrial respiration and fatty acid oxidation (FAO) in the skeletal muscle. Moreover, ncls strongly activates AMPK signaling specifically in the skeletal muscle. The beneficial effects of ncls treatment in fat clearance and AMPK activation were faithfully reproduced in vitro in cultured murine and human primary myotubes. Mechanistically, ncls increases cellular cAMP concentration and ADP/ATP ratio, which further lead to the activation of AMPK signaling. Blocking AMPK signaling through a specific inhibitor significantly reduces FAO in myotubes. Finally, ncls also enhances mitochondrial membrane potential and reduces the formation of reactive oxygen species in cultured myotubes.


Subject(s)
Amaryllidaceae Alkaloids/pharmacology , Amaryllidaceae Alkaloids/therapeutic use , Diet/adverse effects , Muscle, Skeletal/metabolism , Obesity/drug therapy , Obesity/metabolism , Phenanthridines/pharmacology , Phenanthridines/therapeutic use , AMP-Activated Protein Kinases/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Biomarkers/metabolism , Cell Respiration/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Diet, High-Fat , Energy Metabolism/drug effects , Enzyme Activation/drug effects , Fatty Acids/metabolism , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Slow-Twitch/drug effects , Muscle Fibers, Slow-Twitch/metabolism , Muscle, Skeletal/drug effects , Oxidation-Reduction/drug effects , Physical Conditioning, Animal , Protective Agents/pharmacology , Protective Agents/therapeutic use , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
4.
PLoS Genet ; 12(12): e1006474, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27923061

ABSTRACT

Increasing energy expenditure through brown adipocyte recruitment is a promising approach to combat obesity. We report here the comprehensive profiling of the epigenome and transcriptome throughout the lineage commitment and differentiation of C3H10T1/2 mesenchymal stem cell line into brown adipocytes. Through direct comparison to datasets from differentiating white adipocytes, we systematically identify stage- and lineage-specific coding genes, lncRNAs and microRNAs. Utilizing chromatin state maps, we also define stage- and lineage-specific enhancers, including super-enhancers, and their associated transcription factor binding motifs and genes. Through these analyses, we found that in brown adipocytes, brown lineage-specific genes are pre-marked by both H3K4me1 and H3K27me3, and the removal of H3K27me3 at the late stage is necessary but not sufficient to promote brown gene expression, while the pre-deposition of H3K4me1 plays an essential role in poising the brown genes for expression in mature brown cells. Moreover, we identify SOX13 as part of a p38 MAPK dependent transcriptional response mediating early brown cell lineage commitment. We also identify and subsequently validate PIM1, SIX1 and RREB1 as novel regulators promoting brown adipogenesis. Finally, we show that SIX1 binds to adipogenic and brown marker genes and interacts with C/EBPα, C/EBPß and EBF2, suggesting their functional cooperation during adipogenesis.


Subject(s)
Adipogenesis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Proteins/genetics , Homeodomain Proteins/genetics , Obesity/genetics , Adipose Tissue, Brown/growth & development , Adipose Tissue, Brown/metabolism , Animals , Autoantigens/genetics , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Proteins/biosynthesis , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Energy Metabolism/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mesenchymal Stem Cells , Mice , Obesity/metabolism , Obesity/pathology , RNA, Long Noncoding/biosynthesis , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcriptome/genetics
5.
Int J Mol Sci ; 19(6)2018 06 12.
Article in English | MEDLINE | ID: mdl-29895749

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear receptor superfamily and they are essential regulators of cell differentiation, tissue development, and energy metabolism. Given their central roles in sensing the cellular metabolic state and controlling metabolic homeostasis, PPARs became important targets of drug development for the management of metabolic disorders. The function of PPARs is mainly regulated through ligand binding, which induces structural changes, further affecting the interactions with co-activators or co-repressors to stimulate or inhibit their functions. In addition, PPAR functions are also regulated by various Post-translational modifications (PTMs). These PTMs include phosphorylation, SUMOylation, ubiquitination, acetylation, and O-GlcNAcylation, which are found at numerous modification sites. The addition of these PTMs has a wide spectrum of consequences on protein stability, transactivation function, and co-factor interaction. Moreover, certain PTMs in PPAR proteins have been associated with the status of metabolic diseases. In this review, we summarize the PTMs found on the three PPAR isoforms PPARα, PPARß/δ, and PPARγ, and their corresponding modifying enzymes. We also discuss the functional roles of these PTMs in regulating metabolic homeostasis and provide a perspective for future research in this intriguing field.


Subject(s)
Peroxisome Proliferator-Activated Receptors/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Humans , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR delta/genetics , PPAR delta/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , PPAR-beta/genetics , PPAR-beta/metabolism , Peroxisome Proliferator-Activated Receptors/genetics , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology , Receptors, Cytoplasmic and Nuclear/genetics
6.
EMBO J ; 32(24): 3176-91, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24240174

ABSTRACT

The histone deacetylases HDAC1 and HDAC2 remove acetyl moieties from lysine residues of histones and other proteins and are important regulators of gene expression. By deleting different combinations of Hdac1 and Hdac2 alleles in the epidermis, we reveal a dosage-dependent effect of HDAC1/HDAC2 activity on epidermal proliferation and differentiation. Conditional ablation of either HDAC1 or HDAC2 in the epidermis leads to no obvious phenotype due to compensation by the upregulated paralogue. Strikingly, deletion of a single Hdac2 allele in HDAC1 knockout mice results in severe epidermal defects, including alopecia, hyperkeratosis, hyperproliferation and spontaneous tumour formation. These mice display impaired Sin3A co-repressor complex function, increased levels of c-Myc protein, p53 expression and apoptosis in hair follicles (HFs) and misregulation of HF bulge stem cells. Surprisingly, ablation of HDAC1 but not HDAC2 in a skin tumour model leads to accelerated tumour development. Our data reveal a crucial function of HDAC1/HDAC2 in the control of lineage specificity and a novel role of HDAC1 as a tumour suppressor in the epidermis.


Subject(s)
Epidermis/growth & development , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Skin Neoplasms/genetics , Alopecia/genetics , Animals , Apoptosis/genetics , Cell Lineage , Co-Repressor Proteins , Disease Models, Animal , Epidermis/enzymology , Epidermis/pathology , Gene Expression Regulation , Genes, Tumor Suppressor , Genes, p53 , Hair Follicle/pathology , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Keratosis/genetics , Keratosis/pathology , Mice , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Skin Neoplasms/pathology
8.
Nucleic Acids Res ; 43(6): e35, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25223787

ABSTRACT

Next-generation sequencing has been widely used for the genome-wide profiling of histone modifications, transcription factor binding and gene expression through chromatin immunoprecipitated DNA sequencing (ChIP-seq) and cDNA sequencing (RNA-seq). Here, we describe a versatile library construction method that can be applied to both ChIP-seq and RNA-seq on the widely used Illumina platforms. Standard methods for ChIP-seq library construction require nanograms of starting DNA, substantially limiting its application to rare cell types or limited clinical samples. By minimizing the DNA purification steps that cause major sample loss, our method achieved a high sensitivity in ChIP-seq library preparation. Using this method, we achieved the following: (i) generated high-quality epigenomic and transcription factor-binding maps using ChIP-seq for murine adipocytes; (ii) successfully prepared a ChIP-seq library from as little as 25 pg of starting DNA; (iii) achieved paired-end sequencing of the ChIP-seq libraries; (iv) systematically profiled gene expression dynamics during murine adipogenesis using RNA-seq and (v) preserved the strand specificity of the transcripts in RNA-seq. Given its sensitivity and versatility in both double-stranded and single-stranded DNA library construction, this method has wide applications in genomic, epigenomic, transcriptomic and interactomic studies.


Subject(s)
Gene Library , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, RNA/methods , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/genetics , Animals , Chromatin Immunoprecipitation/methods , DNA/genetics , DNA/isolation & purification , Mice , RNA/genetics , RNA/isolation & purification , Transcriptome
9.
EMBO J ; 29(23): 3992-4007, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20967026

ABSTRACT

Histone deacetylase (HDAC) inhibitors induce cell cycle arrest, differentiation or apoptosis in tumour cells and are, therefore, promising anti-cancer reagents. However, the specific HDAC isoforms that mediate these effects are not yet identified. To explore the role of HDAC1 in tumourigenesis and tumour proliferation, we established an experimental teratoma model using wild-type and HDAC1-deficient embryonic stem cells. HDAC1-deficient teratomas showed no significant difference in size compared with wild-type teratomas. Surprisingly, loss of HDAC1 was not only linked to increased apoptosis, but also to significantly enhanced proliferation. Epithelial structures showed reduced differentiation as monitored by Oct3/4 expression and changed E-cadherin localization and displayed up-regulated expression of SNAIL1, a regulator of epithelial cell plasticity. Increased levels of the transcriptional regulator SNAIL1 are crucial for enhanced proliferation and reduced differentiation of HDAC1-deficient teratoma. Importantly, the analysis of human teratomas revealed a similar link between loss of HDAC1 and enhanced tumour malignancy. These findings reveal a novel role for HDAC1 in the control of tumour proliferation and identify HDAC1 as potential marker for benign teratomas.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation, Neoplastic , Histone Deacetylase 1/genetics , Teratoma/enzymology , Animals , Apoptosis , Cadherins/genetics , Carcinoma, Embryonal/enzymology , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/pathology , Histone Deacetylase 1/metabolism , Humans , Mice , Octamer Transcription Factor-3/genetics , Phenotype , Snail Family Transcription Factors , Teratoma/genetics , Teratoma/pathology , Transcription Factors/genetics
10.
PLoS Genet ; 6(4): e1000927, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20442873

ABSTRACT

Large fractions of eukaryotic genomes contain repetitive sequences of which the vast majority is derived from transposable elements (TEs). In order to inactivate those potentially harmful elements, host organisms silence TEs via methylation of transposon DNA and packaging into chromatin associated with repressive histone marks. The contribution of individual histone modifications in this process is not completely resolved. Therefore, we aimed to define the role of reversible histone acetylation, a modification commonly associated with transcriptional activity, in transcriptional regulation of murine TEs. We surveyed histone acetylation patterns and expression levels of ten different murine TEs in mouse fibroblasts with altered histone acetylation levels, which was achieved via chemical HDAC inhibition with trichostatin A (TSA), or genetic inactivation of the major deacetylase HDAC1. We found that one LTR retrotransposon family encompassing virus-like 30S elements (VL30) showed significant histone H3 hyperacetylation and strong transcriptional activation in response to TSA treatment. Analysis of VL30 transcripts revealed that increased VL30 transcription is due to enhanced expression of a limited number of genomic elements, with one locus being particularly responsive to HDAC inhibition. Importantly, transcriptional induction of VL30 was entirely dependent on the activation of MAP kinase pathways, resulting in serine 10 phosphorylation at histone H3. Stimulation of MAP kinase cascades together with HDAC inhibition led to simultaneous phosphorylation and acetylation (phosphoacetylation) of histone H3 at the VL30 regulatory region. The presence of the phosphoacetylation mark at VL30 LTRs was linked with full transcriptional activation of the mobile element. Our data indicate that the activity of different TEs is controlled by distinct chromatin modifications. We show that activation of a specific mobile element is linked to a dual epigenetic mark and propose a model whereby phosphoacetylation of histone H3 is crucial for full transcriptional activation of VL30 elements.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Histones/metabolism , Retroelements/genetics , Acetylation , Animals , Chromatin , Embryo, Mammalian/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Mice , Phosphorylation
11.
EBioMedicine ; 94: 104692, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37451904

ABSTRACT

BACKGROUND: People with Down syndrome (DS) show clinical signs of accelerated ageing. Causative mechanisms remain unknown and hypotheses range from the (essentially untreatable) amplified-chromosomal-instability explanation, to potential actions of individual supernumerary chromosome-21 genes. The latter explanation could open a route to therapeutic amelioration if the specific over-acting genes could be identified and their action toned-down. METHODS: Biological age was estimated through patterns of sugar molecules attached to plasma immunoglobulin-G (IgG-glycans, an established "biological-ageing-clock") in n = 246 individuals with DS from three European populations, clinically characterised for the presence of co-morbidities, and compared to n = 256 age-, sex- and demography-matched healthy controls. Isogenic human induced pluripotent stem cell (hiPSCs) models of full and partial trisomy-21 with CRISPR-Cas9 gene editing and two kinase inhibitors were studied prior and after differentiation to cerebral organoids. FINDINGS: Biological age in adults with DS is (on average) 18.4-19.1 years older than in chronological-age-matched controls independent of co-morbidities, and this shift remains constant throughout lifespan. Changes are detectable from early childhood, and do not require a supernumerary chromosome, but are seen in segmental duplication of only 31 genes, along with increased DNA damage and decreased levels of LaminB1 in nucleated blood cells. We demonstrate that these cell-autonomous phenotypes can be gene-dose-modelled and pharmacologically corrected in hiPSCs and derived cerebral organoids. Using isogenic hiPSC models we show that chromosome-21 gene DYRK1A overdose is sufficient and necessary to cause excess unrepaired DNA damage. INTERPRETATION: Explanation of hitherto observed accelerated ageing in DS as a developmental progeroid syndrome driven by DYRK1A overdose provides a target for early pharmacological preventative intervention strategies. FUNDING: Main funding came from the "Research Cooperability" Program of the Croatian Science Foundation funded by the European Union from the European Social Fund under the Operational Programme Efficient Human Resources 2014-2020, Project PZS-2019-02-4277, and the Wellcome Trust Grants 098330/Z/12/Z and 217199/Z/19/Z (UK). All other funding is described in details in the "Acknowledgements".


Subject(s)
Down Syndrome , Induced Pluripotent Stem Cells , Adult , Humans , Aging , Cell Differentiation , Down Syndrome/genetics , Dyrk Kinases
12.
Plant J ; 67(3): 420-33, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21481027

ABSTRACT

DNA double-strand breaks (DSBs) pose one of the most severe threats to genome integrity, potentially leading to cell death. After detection of a DSB, the DNA damage and repair response is initiated and the DSB is repaired by non-homologous end joining and/or homologous recombination. Many components of these processes are still unknown in Arabidopsis thaliana. In this work, we characterized γ-irradiation and mitomycin C induced 1 (GMI1), a member of the SMC-hinge domain-containing protein family. RT-PCR analysis and promoter-GUS fusion studies showed that γ-irradiation, the radio-mimetic drug bleocin, and the DNA cross-linking agent mitomycin C strongly enhance GMI1 expression particularly in meristematic tissues. The induction of GMI1 by γ-irradiation depends on the signalling kinase Ataxia telangiectasia-mutated (ATM) but not on ATM and Rad3-related (ATR). Epistasis analysis of single and double mutants demonstrated that ATM acts upstream of GMI1 while the atr gmi1-2 double mutant was more sensitive than the respective single mutants. Comet assay revealed a reduced rate of DNA double-strand break repair in gmi1 mutants during the early recovery phase after exposure to bleocin. Moreover, the rate of homologous recombination of a reporter construct was strongly reduced in gmi1 mutant plants upon exposure to bleocin or mitomycin C. GMI1 is the first member of its protein family known to be involved in DNA repair.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/genetics , Chromosomes, Plant/metabolism , DNA, Plant/metabolism , Arabidopsis/drug effects , Arabidopsis/metabolism , Arabidopsis/radiation effects , Arabidopsis Proteins/genetics , Cloning, Molecular , Comet Assay , DNA Breaks, Double-Stranded , DNA Repair , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , DNA, Plant/genetics , Flowers/drug effects , Flowers/metabolism , Flowers/radiation effects , Gene Expression Regulation, Plant , Gene Fusion , Meristem/drug effects , Meristem/metabolism , Meristem/radiation effects , Microarray Analysis , Mitomycin/pharmacology , Mutagenesis, Insertional , Recombination, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Seedlings/drug effects , Seedlings/genetics , Transcription, Genetic
13.
Article in English | MEDLINE | ID: mdl-32174890

ABSTRACT

Over the past decade, the increasing prevalence of obesity and its associated metabolic disorders constitutes one of the most concerning healthcare issues for countries worldwide. In an effort to curb the increased mortality and morbidity derived from the obesity epidemic, various therapeutic strategies have been developed by researchers. In the recent years, advances in the field of adipocyte biology have revealed that the thermogenic adipose tissue holds great potential in ameliorating metabolic disorders. Additionally, epigenetic research has shed light on the effects of histone acetylation on adipogenesis and thermogenesis, thereby establishing the essential roles which histone acetyltransferases (HATs) and histone deacetylases (HDACs) play in metabolism and systemic energy homeostasis. In regard to the therapeutic potential of thermogenic adipocytes for the treatment of metabolic diseases, herein, we describe the current state of knowledge of the regulation of thermogenic adipocyte differentiation and adaptive thermogenesis through histone acetylation. Furthermore, we highlight how different HATs and HDACs maintain the epigenetic transcriptional network to mediate the pathogenesis of various metabolic comorbidities. Finally, we provide insights into recent advances of the potential therapeutic applications and development of HAT and HDAC inhibitors to alleviate these pathological conditions.


Subject(s)
Adaptation, Physiological , Adipocytes/cytology , Adipogenesis , Cell Differentiation , Histones/chemistry , Thermogenesis , Acetylation , Adipocytes/physiology , Animals , Humans
14.
PLoS One ; 12(3): e0174761, 2017.
Article in English | MEDLINE | ID: mdl-28358928

ABSTRACT

Adipose tissues constitute an important component of metabolism, the dysfunction of which can cause obesity and type II diabetes. Here we show that differentiation of white and brown adipocytes requires Deleted in Liver Cancer 1 (DLC1), a Rho GTPase Activating Protein (RhoGAP) previously studied for its function in liver cancer. We identified Dlc1 as a super-enhancer associated gene in both white and brown adipocytes through analyzing the genome-wide binding profiles of PPARγ, the master regulator of adipogenesis. We further observed that Dlc1 expression increases during differentiation, and knockdown of Dlc1 by siRNA in white adipocytes reduces the formation of lipid droplets and the expression of fat marker genes. Moreover, knockdown of Dlc1 in brown adipocytes reduces expression of brown fat-specific genes and diminishes mitochondrial respiration. Dlc1-/- knockout mouse embryonic fibroblasts show a complete inability to differentiate into adipocytes, but this phenotype can be rescued by inhibitors of Rho-associated kinase (ROCK) and filamentous actin (F-actin), suggesting the involvement of Rho pathway in DLC1-regulated adipocyte differentiation. Furthermore, PPARγ binds to the promoter of Dlc1 gene to regulate its expression during both white and brown adipocyte differentiation. These results identify DLC1 as an activator of white and brown adipocyte differentiation, and provide a molecular link between PPARγ and Rho pathways.


Subject(s)
Adipocytes, Brown/cytology , Adipocytes, Brown/metabolism , Adipocytes, White/cytology , Adipocytes, White/metabolism , GTPase-Activating Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Adipogenesis/genetics , Adipogenesis/physiology , Blotting, Western , Calorimetry, Indirect , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Chromatin Immunoprecipitation , GTPase-Activating Proteins/genetics , Humans , Oxygen Consumption/genetics , Oxygen Consumption/physiology , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Small Interfering/genetics , Tumor Suppressor Proteins/genetics
15.
PLoS One ; 11(9): e0162528, 2016.
Article in English | MEDLINE | ID: mdl-27606599

ABSTRACT

Lysine acetylation is an important post-translational modification in cell signaling. In acetylome studies, a high-quality pan-acetyl-lysine antibody is key to successful enrichment of acetylated peptides for subsequent mass spectrometry analysis. Here we show an alternative method to generate polyclonal pan-acetyl-lysine antibodies using a synthesized random library of acetylated peptides as the antigen. Our antibodies are tested to be specific for acetyl-lysine peptides/proteins via ELISA and dot blot. When pooled, five of our antibodies show broad reactivity to acetyl-lysine peptides, complementing a commercial antibody in terms of peptide coverage. The consensus sequence of peptides bound by our antibody cocktail differs slightly from that of the commercial antibody. Lastly, our antibodies are tested in a proof-of-concept to analyze the acetylome of HEK293 cells. In total we identified 1557 acetylated peptides from 416 proteins. We thus demonstrated that our antibodies are well-qualified for acetylome studies and can complement existing commercial antibodies.


Subject(s)
Antibodies/metabolism , Lysine/metabolism , 3T3-L1 Cells , Acetylation , Amino Acid Motifs , Animals , Chromatography, Liquid , Consensus Sequence , Enzyme-Linked Immunosorbent Assay , Gene Ontology , HEK293 Cells , Humans , Immunoassay , Male , Metabolome , Mice , Peptides/chemistry , Peptides/metabolism , Rabbits , Reproducibility of Results , Tandem Mass Spectrometry
16.
PLoS One ; 10(7): e0133448, 2015.
Article in English | MEDLINE | ID: mdl-26177453

ABSTRACT

SIRT1 plays a key role in maintaining metabolic homeostasis in mammals by directly modulating the activities of various transcription factors and metabolic enzymes through lysine deacetylation. White adipose tissue plays a key role in lipid storage and metabolism. To identify novel molecular targets of SIRT1 in fat cells, we used a non-biased proteomic approach. We identified a number of proteins whose acetylation status was significantly affected by SIRT1 modulator treatment in 3T3-L1 adipocytes. Among them, ATP6V1B2, a subunit of the vacuolar (H+)-ATPase, was further shown to be associated with SIRT1 by co-immunoprecipitation assay. Moreover, SIRT1 deacetylates ATP6V1B2 in vitro and in vivo. Taken together, our study demonstrates that ATP6V1B2 is a molecular target of SIRT1 in fat cells and the role of SIRT1 and ATP6V1B2 acetylation in the vacuolar (H+)-ATPase function warrants further investigation.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Cell Differentiation , Sirtuin 1/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , 3T3-L1 Cells , Acetylation , Animals , HEK293 Cells , Humans , Mice , Protein Binding , Proteomics
17.
Cell Cycle ; 11(23): 4310-22, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23085542

ABSTRACT

Histone modifications and their modifying enzymes are fundamentally involved in the epigenetic regulation of adipogenesis. This study aimed to define the roles of various histone modifications and their "division of labor" in fat cell differentiation. To achieve these goals, we examined the distribution patterns of eight core histone modifications at five key adipogenic regulatory genes, Pref-1, C/EBPß, C/EBPα, PPARγ2 and aP2, during the adipogenesis of C3H 10T1/2 mouse mesenchymal stem cells (MSCs) and 3T3-L1 preadipocytes. We found that the examined histone modifications are globally stable throughout adipogenesis but show distinct and highly dynamic distribution patterns at specific genes. For example, the Pref-1 gene has lower levels of active chromatin markers and significantly higher H3 K27 tri-methylation in MSCs compared with committed preadipocytes; the C/EBPß gene is enriched in active chromatin markers at its 3'-UTR; the C/EBPα gene is predominantly marked by H3 K27 tri-methylation in adipogenic precursor cells, and this repressive marker decreases dramatically upon induction; the PPARγ2 and aP2 genes show increased histone acetylation on both H3 and H4 tails during adipogenesis. Further functional studies revealed that the decreased level of H3 K27 tri-methylation leads to de-repression of Pref-1 gene, while the increased level of histone acetylation activates the transcription of PPARγ2 and aP2 genes. Moreover, the active histone modification-marked 3'-UTR of C/EBPß gene was demonstrated as a strong enhancer element by luciferase assay. Our results indicate that histone modifications are gene-specific at adipogenic regulator genes, and they play distinct roles in regulating the transcriptional network during adipogenesis.


Subject(s)
Adipogenesis/genetics , Histones/metabolism , 3' Untranslated Regions , 3T3-L1 Cells , Acetylation , Adipocytes/cytology , Adipocytes/metabolism , Animals , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Calcium-Binding Proteins , Cell Line , Epigenesis, Genetic , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Methylation , Mice , PPAR gamma/genetics , PPAR gamma/metabolism
18.
Mol Cell Biol ; 30(5): 1171-81, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20028735

ABSTRACT

Histone deacetylases (HDACs) are chromatin-modifying enzymes that are involved in the regulation of proliferation, differentiation and development. HDAC inhibitors induce cell cycle arrest, differentiation, or apoptosis in tumor cells and are therefore promising antitumor agents. Numerous genes were found to be deregulated upon HDAC inhibitor treatment; however, the relevant target enzymes are still unidentified. HDAC1 is required for mouse development and unrestricted proliferation of embryonic stem cells. We show here that HDAC1 reversibly regulates cellular proliferation and represses the cyclin-dependent kinase inhibitor p21 in embryonic stem cells. Disruption of the p21 gene rescues the proliferation phenotype of HDAC1(-/-) embryonic stem cells but not the embryonic lethality of HDAC1(-/-) mice. In the absence of HDAC1, mouse embryonic fibroblasts scarcely undergo spontaneous immortalization and display increased p21 expression. Chromatin immunoprecipitation assays demonstrate a direct regulation of the p21 gene by HDAC1 in mouse embryonic fibroblasts. Transformation with simian virus 40 large T antigen or ablation of p21 restores normal immortalization of primary HDAC1(-/-) fibroblasts. Our data demonstrate that repression of the p21 gene is crucial for HDAC1-mediated control of proliferation and immortalization. HDAC1 might therefore be one of the relevant targets for HDAC inhibitors as anticancer drugs.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Histone Deacetylase 1/metabolism , Animals , Antigens, Polyomavirus Transforming/genetics , Cell Transformation, Viral , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Gene Expression , Histone Deacetylase 1/deficiency , Histone Deacetylase 1/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Phenotype , Repressor Proteins/genetics
19.
Int J Dev Biol ; 53(2-3): 275-89, 2009.
Article in English | MEDLINE | ID: mdl-19412887

ABSTRACT

During development from the fertilized egg to a multicellular organism, cell fate decisions have to be taken and cell lineage or tissue-specific gene expression patterns are created and maintained. These alterations in gene expression occur in the context of chromatin structure and are controlled by chromatin modifying enzymes. Gene disruption studies in different genetic systems have shown an essential role of various histone deacetylases (HDACs) during early development and cellular differentiation. In this review, we focus on the functions of the class I enzymes HDAC1 and HDAC2 during development in different organisms and summarise the current knowledge about their involvement in neurogenesis, myogenesis, haematopoiesis and epithelial cell differentiation.


Subject(s)
Cell Differentiation/physiology , Embryonic Development/physiology , Histone Deacetylases/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Histone Deacetylase 1 , Histone Deacetylase 2 , Histone Deacetylases/classification , Histone Deacetylases/genetics , Mice , Models, Biological , Phylogeny , Repressor Proteins/classification , Repressor Proteins/genetics
20.
EMBO J ; 21(11): 2672-81, 2002 Jun 03.
Article in English | MEDLINE | ID: mdl-12032080

ABSTRACT

Histone deacetylases (HDACs) modulate chromatin structure and transcription, but little is known about their function in mammalian development. HDAC1 was implicated previously in the repression of genes required for cell proliferation and differentiation. Here we show that targeted disruption of both HDAC1 alleles results in embryonic lethality before E10.5 due to severe proliferation defects and retardation in development. HDAC1-deficient embryonic stem cells show reduced proliferation rates, which correlate with decreased cyclin-associated kinase activities and elevated levels of the cyclin-dependent kinase inhibitors p21(WAF1/CIP1) and p27(KIP1). Similarly, expression of p21 and p27 is up-regulated in HDAC1-null embryos. In addition, loss of HDAC1 leads to significantly reduced overall deacetylase activity, hyperacetylation of a subset of histones H3 and H4 and concomitant changes in other histone modifications. The expression of HDAC2 and HDAC3 is induced in HDAC1-deficient cells, but cannot compensate for loss of the enzyme, suggesting a unique function for HDAC1. Our study provides the first evidence that a histone deacetylase is essential for unrestricted cell proliferation by repressing the expression of selective cell cycle inhibitors.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/physiology , Alleles , Animals , Blotting, Southern , Blotting, Western , Cell Cycle Proteins/metabolism , Cell Division , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/metabolism , Exons , Histone Deacetylase 1 , Histones/metabolism , In Situ Hybridization , In Situ Nick-End Labeling , Mice , Microscopy, Fluorescence , Models, Genetic , Phenotype , Precipitin Tests , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Suppressor Proteins/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL