Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 81
1.
Proc Natl Acad Sci U S A ; 120(31): e2308750120, 2023 08.
Article En | MEDLINE | ID: mdl-37487068

Adipose tissue is central to regulation of energy homeostasis. Adaptive thermogenesis, which relies on mitochondrial oxidative phosphorylation (Ox-Phos), dissipates energy to counteract obesity. On the other hand, chronic inflammation in adipose tissue is linked to type 2 diabetes and obesity. Here, we show that nuclear factor I-A (NFIA), a transcriptional regulator of brown and beige adipocytes, improves glucose homeostasis by upregulation of Ox-Phos and reciprocal downregulation of inflammation. Mice with transgenic expression of NFIA in adipocytes exhibited improved glucose tolerance and limited weight gain. NFIA up-regulates Ox-Phos and brown-fat-specific genes by enhancer activation that involves facilitated genomic binding of PPARγ. In contrast, NFIA in adipocytes, but not in macrophages, down-regulates proinflammatory cytokine genes to ameliorate adipose tissue inflammation. NFIA binds to regulatory region of the Ccl2 gene, which encodes proinflammatory cytokine MCP-1 (monocyte chemoattractant protein-1), to down-regulate its transcription. CCL2 expression was negatively correlated with NFIA expression in human adipose tissue. These results reveal the beneficial effect of NFIA on glucose and body weight homeostasis and also highlight previously unappreciated role of NFIA in suppressing adipose tissue inflammation.


Diabetes Mellitus, Type 2 , NFI Transcription Factors , Humans , Animals , Mice , Adipocytes , Homeostasis , Inflammation , Adipose Tissue, Brown , Cytokines
2.
Nat Commun ; 13(1): 4501, 2022 08 30.
Article En | MEDLINE | ID: mdl-36042201

KMT2A-rearranged infant acute lymphoblastic leukemia (ALL) represents the most refractory type of childhood leukemia. To uncover the molecular heterogeneity of this disease, we perform RNA sequencing, methylation array analysis, whole exome and targeted deep sequencing on 84 infants with KMT2A-rearranged leukemia. Our multi-omics clustering followed by single-sample and single-cell inference of hematopoietic differentiation establishes five robust integrative clusters (ICs) with different master transcription factors, fusion partners and corresponding stages of B-lymphopoietic and early hemato-endothelial development: IRX-type differentiated (IC1), IRX-type undifferentiated (IC2), HOXA-type MLLT1 (IC3), HOXA-type MLLT3 (IC4), and HOXA-type AFF1 (IC5). Importantly, our deep mutational analysis reveals that the number of RAS pathway mutations predicts prognosis and that the most refractory subgroup of IC2 possesses 100% frequency and the heaviest burden of RAS pathway mutations. Our findings highlight the previously under-appreciated intra- and inter-patient heterogeneity of KMT2A-rearranged infant ALL and provide a rationale for the future development of genomics-guided risk stratification and individualized therapy.


Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Gene Fusion , Humans , Infant , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/genetics
3.
iScience ; 25(8): 104729, 2022 Aug 19.
Article En | MEDLINE | ID: mdl-35874098

Thermogenic brown and beige adipocytes counteract obesity by enhancing energy dissipation via uncoupling protein-1 (Ucp1). However, the effect of genetic variation on these cells, a major source of disease susceptibility, has been less well studied. Here we examined beige adipocytes from obesity-prone C57BL/6J (B6) and obesity-resistant 129X1/SvJ (129) mouse strains and identified a cis-regulatory variant rs47238345 that is responsible for differential Ucp1 expression. The alternative T allele of rs47238345 at the Ucp1 -12kb enhancer in 129 facilitates the allele-specific binding of nuclear factor I-A (NFIA) to mediate allele-specific enhancer-promoter interaction and Ucp1 transcription. Furthermore, CRISPR-Cas9/Cpf1-mediated single nucleotide polymorphism (SNP) editing of rs47238345 resulted in increased Ucp1 expression. We also identified Lim homeobox protein 8 (Lhx8), whose expression is higher in 129 than in B6, as a trans-acting regulator of Ucp1 in mice and humans. These results demonstrate the cis- and trans-acting effects of genetic variation on Ucp1 expression that underlie phenotypic diversity.

4.
JTO Clin Res Rep ; 3(2): 100277, 2022 Feb.
Article En | MEDLINE | ID: mdl-35199052

We analyzed an EGFR-mutated lung cancer with a pathologic diagnosis of combined large cell neuroendocrine carcinoma with mixed adenocarcinoma subtypes. Targeted next-generation sequencing of each component suggested that mutations in RB1, TP53, and SMAD4 and apparent loss of heterozygosity of TP53 and SMAD4 accompanied the transition of different adenocarcinoma subtypes. Additional gene mutations including PTEN, MST1R, and PIK3CA were noted during transdifferentiation from acinar adenocarcinoma to large cell neuroendocrine carcinoma. Combined DNA and RNA analysis using Todai OncoPanel revealed that transdifferentiation to different pathologic subtypes occurred in a single tumor through the accumulation of gene mutations.

5.
J Biochem ; 171(4): 399-410, 2022 Mar 31.
Article En | MEDLINE | ID: mdl-34908107

Smad proteins transduce signals downstream of transforming growth factor-ß (TGF-ß) and are one of the factors that regulate the expression of genes related to diseases affecting the skin. In the present study, we identified MAB21L4, also known as male abnormal 21 like 4 or C2orf54, as the most up-regulated targets of TGF-ß and Smad3 in differentiated human progenitor epidermal keratinocytes using chromatin immunoprecipitation sequencing (ChIP-seq) and RNA sequencing (RNA-seq). We found that TGF-ß induced expression of the barrier protein involucrin (encoded by the IVL gene). Transcriptional activity of the IVL promoter induced by TGF-ß was inhibited by MAB21L4 siRNAs. Further analysis revealed that MAB21L4 siRNAs also down-regulated the expression of several target genes of TGF-ß. MAB21L4 protein was located mainly in the cytosol, where it was physically bound to Smad3 and a transcriptional corepressor c-Ski. siRNAs for MAB21L4 did not inhibit the binding of Smad3 to their target genomic regions but down-regulated the acetylation of histone H3 lys 27 (H3K27ac), an active histone mark, near the Smad3 binding regions. These findings suggest that TGF-ß-induced MAB21L4 up-regulates the gene expression induced by TGF-ß, possibly through the inhibition of c-Ski via physical interaction in the cytosol.


Intracellular Signaling Peptides and Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta , Humans , Keratinocytes/metabolism , Male , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
6.
Cancer Sci ; 113(3): 940-949, 2022 Mar.
Article En | MEDLINE | ID: mdl-34897916

The activation of RIG-I-like receptor (RLR) signaling in cancer cells is widely recognized as a critical cancer therapy method. The expected mechanism of RLR ligand-mediated cancer therapy involves the promotion of cancer cell death and strong induction of interferon (IFN)-ß that affects the tumor microenvironment. We have recently shown that activation of RLR signaling in triple-negative breast cancer cells (TNBC) attenuates transforming growth factor-ß (TGF-ß) signaling, which partly contributes to the promotion of cancer cell pyroptosis. However, the consequences of suppression of TGF-ß signaling by RLR ligands with respect to IFN-ß-mediated tumor suppression are not well characterized. This study showed that transfection of a typical RLR ligand polyI:C in cancer cells produces significant levels of IFN-ß, which inhibits the growth of the surrounding cancer cells. In addition, IFN-ß-induced cell cycle arrest in surrounding cancer cells was inhibited by the expression of constitutively active Smad3. Constitutively active Smad3 suppresses IFN-ß expression through the alleviation of IFN regulatory factor 3 binding to the canonical target genes, as suggested by ChIP sequencing analysis. Based on these findings, a new facet of the protumorigenic function of TGF-ß that suppresses IFN-ß expression is suggested when RLR-mediated cancer treatment is used in TNBC.


Interferon-beta/metabolism , Poly I-C/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Triple Negative Breast Neoplasms/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factor-3/metabolism , Poly I-C/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transfection , Transforming Growth Factor beta/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/drug effects
7.
Nat Commun ; 12(1): 7045, 2021 12 02.
Article En | MEDLINE | ID: mdl-34857762

Enhancer activation is essential for cell-type specific gene expression during cellular differentiation, however, how enhancers transition from a hypoacetylated "primed" state to a hyperacetylated-active state is incompletely understood. Here, we show SET domain-containing 5 (SETD5) forms a complex with NCoR-HDAC3 co-repressor that prevents histone acetylation of enhancers for two master adipogenic regulatory genes Cebpa and Pparg early during adipogenesis. The loss of SETD5 from the complex is followed by enhancer hyperacetylation. SETD5 protein levels were transiently increased and rapidly degraded prior to enhancer activation providing a mechanism for the loss of SETD5 during the transition. We show that induction of the CDC20 co-activator of the ubiquitin ligase leads to APC/C mediated degradation of SETD5 during the transition and this operates as a molecular switch that facilitates adipogenesis.


Adipogenesis/genetics , CCAAT-Enhancer-Binding Proteins/genetics , Histone Deacetylases/genetics , Methyltransferases/genetics , Nuclear Receptor Co-Repressor 1/genetics , PPAR gamma/genetics , 3T3-L1 Cells , Acetylation , Anaphase-Promoting Complex-Cyclosome/genetics , Anaphase-Promoting Complex-Cyclosome/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cdc20 Proteins/genetics , Cdc20 Proteins/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , HEK293 Cells , Histone Deacetylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Methyltransferases/metabolism , Mice , Mice, Nude , Nuclear Receptor Co-Repressor 1/metabolism , PPAR gamma/metabolism , Protein Binding , Proteolysis , Sf9 Cells , Signal Transduction
8.
Cancer Sci ; 112(7): 2855-2869, 2021 Jul.
Article En | MEDLINE | ID: mdl-33970549

Ten-eleven translocation 1 (TET1) is an essential methylcytosine dioxygenase of the DNA demethylation pathway. Despite its dysregulation being known to occur in human cancer, the role of TET1 remains poorly understood. In this study, we report that TET1 promotes cell growth in human liver cancer. The transcriptome analysis of 68 clinical liver samples revealed a subgroup of TET1-upregulated hepatocellular carcinoma (HCC), demonstrating hepatoblast-like gene expression signatures. We performed comprehensive cytosine methylation and hydroxymethylation (5-hmC) profiling and found that 5-hmC was aberrantly deposited preferentially in active enhancers. TET1 knockdown in hepatoma cell lines decreased hmC deposition with cell growth suppression. HMGA2 was highly expressed in a TET1high subgroup of HCC, associated with the hyperhydroxymethylation of its intronic region, marked as histone H3K4-monomethylated, where the H3K27-acetylated active enhancer chromatin state induced interactions with its promoter. Collectively, our findings point to a novel type of epigenetic dysregulation, methylcytosine dioxygenase TET1, which promotes cell proliferation via the ectopic enhancer of its oncogenic targets, HMGA2, in hepatoblast-like HCC.


HMGA2 Protein/genetics , Liver Neoplasms/genetics , Mixed Function Oxygenases/genetics , Neoplasm Proteins/genetics , Proto-Oncogene Proteins/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Chromatin/genetics , Cytosine/metabolism , DNA Methylation , Dioxygenases/metabolism , Epigenesis, Genetic , Gene Expression , Gene Knockdown Techniques , HMGA2 Protein/metabolism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mixed Function Oxygenases/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Up-Regulation
9.
Blood ; 137(1): 75-88, 2021 01 07.
Article En | MEDLINE | ID: mdl-32730594

The pseudokinase Trib1 functions as a myeloid oncogene that recruits the E3 ubiquitin ligase COP1 to C/EBPα and interacts with MEK1 to enhance extracellular signal-regulated kinase (ERK) phosphorylation. A close genetic effect of Trib1 on Hoxa9 has been observed in myeloid leukemogenesis, where Trib1 overexpression significantly accelerates Hoxa9-induced leukemia onset. However, the mechanism underlying how Trib1 functionally modulates Hoxa9 transcription activity is unclear. Herein, we provide evidence that Trib1 modulates Hoxa9-associated super-enhancers. Chromatin immunoprecipitation sequencing analysis identified increased histone H3K27Ac signals at super-enhancers of the Erg, Spns2, Rgl1, and Pik3cd loci, as well as increased messenger RNA expression of these genes. Modification of super-enhancer activity was mostly achieved via the degradation of C/EBPα p42 by Trib1, with a slight contribution from the MEK/ERK pathway. Silencing of Erg abrogated the growth advantage acquired by Trib1 overexpression, indicating that Erg is a critical downstream target of the Trib1/Hoxa9 axis. Moreover, treatment of acute myeloid leukemia (AML) cells with the BRD4 inhibitor JQ1 showed growth inhibition in a Trib1/Erg-dependent manner both in vitro and in vivo. Upregulation of ERG by TRIB1 was also observed in human AML cell lines, suggesting that Trib1 is a potential therapeutic target of Hoxa9-associated AML. Taken together, our study demonstrates a novel mechanism by which Trib1 modulates chromatin and Hoxa9-driven transcription in myeloid leukemogenesis.


Gene Expression Regulation, Leukemic/genetics , Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, Myeloid, Acute/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Disease Progression , Homeodomain Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription, Genetic
10.
PLoS Genet ; 16(9): e1009044, 2020 09.
Article En | MEDLINE | ID: mdl-32991581

The transcription factor nuclear factor I-A (NFIA) is a regulator of brown adipocyte differentiation. Here we show that the C-terminal 17 amino acid residues of NFIA (which we call pro#3 domain) are required for the transcriptional activity of NFIA. Full-length NFIA-but not deletion mutant lacking pro#3 domain-rescued impaired expression of PPARγ, the master transcriptional regulator of adipogenesis and impaired adipocyte differentiation in NFIA-knockout cells. Mechanistically, the ability of NFIA to penetrate chromatin and bind to the crucial Pparg enhancer is mediated through pro#3 domain. However, the deletion mutant still binds to Myod1 enhancer to repress expression of MyoD, the master transcriptional regulator of myogenesis as well as proximally transcribed non-coding RNA called DRReRNA, via competition with KLF5 in terms of enhancer binding, leading to suppression of myogenic gene program. Therefore, the negative effect of NFIA on the myogenic gene program is, at least partly, independent of the positive effect on PPARγ expression and its downstream adipogenic gene program. These results uncover multiple ways of action of NFIA to ensure optimal regulation of brown and beige adipocyte differentiation.


Adipocytes, Beige/cytology , Adipocytes, Brown/cytology , Adipogenesis/physiology , Muscle Development/physiology , NFI Transcription Factors/metabolism , Adipocytes, Beige/physiology , Adipocytes, Brown/physiology , Adipogenesis/genetics , Animals , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Knockout , Muscle Development/genetics , MyoD Protein/genetics , Myogenin/genetics , NFI Transcription Factors/genetics , PPAR gamma/genetics , PPAR gamma/metabolism , Proline , Protein Domains
11.
EMBO J ; 39(7): e103949, 2020 04 01.
Article En | MEDLINE | ID: mdl-32125007

Histone H3 lysine-9 di-methylation (H3K9me2) and lysine-27 tri-methylation (H3K27me3) are linked to repression of gene expression, but the functions of repressive histone methylation dynamics during inflammatory responses remain enigmatic. Here, we report that lysine demethylases 7A (KDM7A) and 6A (UTX) play crucial roles in tumor necrosis factor (TNF)-α signaling in endothelial cells (ECs), where they are regulated by a novel TNF-α-responsive microRNA, miR-3679-5p. TNF-α rapidly induces co-occupancy of KDM7A and UTX at nuclear factor kappa-B (NF-κB)-associated elements in human ECs. KDM7A and UTX demethylate H3K9me2 and H3K27me3, respectively, and are both required for activation of NF-κB-dependent inflammatory genes. Chromosome conformation capture-based methods furthermore uncover increased interactions between TNF-α-induced super enhancers at NF-κB-relevant loci, coinciding with KDM7A and UTX recruitments. Simultaneous pharmacological inhibition of KDM7A and UTX significantly reduces leukocyte adhesion in mice, establishing the biological and potential translational relevance of this mechanism. Collectively, these findings suggest that rapid erasure of repressive histone marks by KDM7A and UTX is essential for NF-κB-dependent regulation of genes that control inflammatory responses of ECs.


Endothelial Cells/immunology , Histone Demethylases/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , MicroRNAs/genetics , Animals , Cell Adhesion , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Histones/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Lysine/metabolism , Male , Methylation , Mice , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
12.
Cancer Sci ; 111(2): 451-466, 2020 Feb.
Article En | MEDLINE | ID: mdl-31834974

The tumor suppressor gene p53 encodes a transcriptional activator that has two transactivation domains (TAD) located in its amino terminus. These two TAD can transactivate genes independently, and at least one TAD is required for p53 transactivation function. The 1st TAD (a.a. 1-40) is essential for the induction of numerous classical p53 target genes, while the second TAD (a.a. 41-61) suffices for tumor suppression, although its precise molecular function remains unclear. In this study, we comprehensively identified the sites to which p53 lacking the 1st TAD (Δ1stTAD-p53) binds, as well as its potential target genes. We found that the binding sequences for Δ1stTAD-p53 are divergent and include not only the canonical p53 consensus binding sequences but also sequences similar to those recognized by a number of other known transcription factors. We identified and analyzed the functions of three Δ1stTAD-p53 target genes, PTP4A1, PLK2 and RPS27L. All three genes were induced by both full-length p53 and Δ1stTAD-p53, and were dependent on the transactivation activity of the 2nd TAD. We also found that two of these, PTP4A1 and PLK2, are endoplasmic reticulum (ER) stress-inducible genes. We found that upon ER stress, PTP4A1 suppresses apoptosis while PLK2 induces apoptosis. These results reveal a novel Δ1stTAD-p53 downstream pathway that is dependent on the transcription activation activity of the 2nd TAD.


Cell Cycle Proteins/genetics , Membrane Proteins/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Tyrosine Phosphatases/genetics , Ribosomal Proteins/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Apoptosis , Binding Sites , Endoplasmic Reticulum Stress , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Oligonucleotide Array Sequence Analysis , Protein Binding , Protein Domains , Transcriptional Activation , Tumor Suppressor Protein p53/genetics
13.
Mol Oncol ; 14(2): 277-293, 2020 02.
Article En | MEDLINE | ID: mdl-31782890

Thyroid transcription factor-1 (TTF-1, encoded by the NKX2-1 gene) is highly expressed in small-cell lung carcinoma (SCLC) and lung adenocarcinoma (LADC), but how its functional roles differ between SCLC and LADC remains to be elucidated. Here, we compared the genome-wide distributions of TTF-1 binding regions and the transcriptional programs regulated by TTF-1 between NCI-H209 (H209), a human SCLC cell line, and NCI-H441 (H441), a human LADC cell line, using chromatin immunoprecipitation-sequencing (ChIP-seq) and RNA-sequencing (RNA-seq). TTF-1 binding regions in H209 and H441 cells differed by 75.0% and E-box motifs were highly enriched exclusively in the TTF-1 binding regions of H209 cells. Transcriptome profiling revealed that TTF-1 is involved in neuroendocrine differentiation in H209 cells. We report that TTF-1 and achaete-scute homolog 1 (ASCL1, also known as ASH1, an E-box binding basic helix-loop-helix transcription factor, and a lineage-survival oncogene of SCLC) are coexpressed and bound to adjacent sites on target genes expressed in SCLC, and cooperatively regulate transcription. Furthermore, TTF-1 regulated expression of the Bcl-2 gene family and showed antiapoptotic function in SCLC. Our findings suggest that TTF-1 promotes SCLC growth and contributes to neuroendocrine and antiapoptotic gene expression by partly coordinating with ASCL1.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Cell Proliferation/genetics , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/genetics , Thyroid Nuclear Factor 1/metabolism , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Cell Line, Tumor , Cell Survival/genetics , Chromatin Immunoprecipitation Sequencing , Gene Expression Regulation, Neoplastic/genetics , Gene Ontology , Genome-Wide Association Study , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Nucleotide Motifs , Prognosis , Protein Binding , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA-Seq , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/mortality , Tissue Array Analysis
14.
Epigenetics Chromatin ; 12(1): 77, 2019 12 19.
Article En | MEDLINE | ID: mdl-31856914

BACKGROUND: Endothelial cells (ECs) make up the innermost layer throughout the entire vasculature. Their phenotypes and physiological functions are initially regulated by developmental signals and extracellular stimuli. The underlying molecular mechanisms responsible for the diverse phenotypes of ECs from different organs are not well understood. RESULTS: To characterize the transcriptomic and epigenomic landscape in the vascular system, we cataloged gene expression and active histone marks in nine types of human ECs (generating 148 genome-wide datasets) and carried out a comprehensive analysis with chromatin interaction data. We developed a robust procedure for comparative epigenome analysis that circumvents variations at the level of the individual and technical noise derived from sample preparation under various conditions. Through this approach, we identified 3765 EC-specific enhancers, some of which were associated with disease-associated genetic variations. We also identified various candidate marker genes for each EC type. We found that the nine EC types can be divided into two subgroups, corresponding to those with upper-body origins and lower-body origins, based on their epigenomic landscape. Epigenomic variations were highly correlated with gene expression patterns, but also provided unique information. Most of the deferentially expressed genes and enhancers were cooperatively enriched in more than one EC type, suggesting that the distinct combinations of multiple genes play key roles in the diverse phenotypes across EC types. Notably, many homeobox genes were differentially expressed across EC types, and their expression was correlated with the relative position of each organ in the body. This reflects the developmental origins of ECs and their roles in angiogenesis, vasculogenesis and wound healing. CONCLUSIONS: This comprehensive analysis of epigenome characterization of EC types reveals diverse transcriptional regulation across human vascular systems. These datasets provide a valuable resource for understanding the vascular system and associated diseases.


Endothelial Cells/metabolism , Epigenome , Gene Expression Regulation , Chromatin/metabolism , Databases, Genetic , Endothelial Cells/cytology , Enhancer Elements, Genetic , Genome-Wide Association Study , Histone Code , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Principal Component Analysis , Promoter Regions, Genetic
15.
J Biol Chem ; 294(42): 15466-15479, 2019 10 18.
Article En | MEDLINE | ID: mdl-31481467

Smad proteins are transcriptional regulators activated by TGF-ß. They are known to bind to two distinct Smad-responsive motifs, namely the Smad-binding element (SBE) (5'-GTCTAGAC-3') and CAGA motifs (5'-AGCCAGACA-3' or 5'-TGTCTGGCT-3'). However, the mechanisms by which these motifs promote Smad activity are not fully elucidated. In this study, we performed DNA CASTing, binding assays, ChIP sequencing, and quantitative RT-PCR to dissect the details of Smad binding and function of the SBE and CAGA motifs. We observed a preference for Smad3 to bind CAGA motifs and Smad4 to bind SBE, and that either one SBE or a triple-CAGA motif forms a cis-acting functional half-unit for Smad-dependent transcription activation; combining two half-units allows efficient activation. Unexpectedly, the extent of Smad binding did not directly correlate with the abilities of Smad-binding sequences to induce gene expression. We found that Smad proteins are more tolerant of single bp mutations in the context of the CAGA motifs, with any mutation in the SBE disrupting function. CAGA and CAGA-like motifs but not SBE are widely distributed among stimulus-dependent Smad2/3-binding sites in normal murine mammary gland epithelial cells, and the number of CAGA and CAGA-like motifs correlates with fold-induction of target gene expression by TGF-ß. These data, demonstrating Smad responsiveness can be tuned by both sequence and number of repeats, provide a compelling explanation for why CAGA motifs are predominantly used for Smad-dependent transcription activation in vivo.


Smad3 Protein/chemistry , Smad3 Protein/metabolism , Smad4 Protein/chemistry , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Amino Acid Motifs , Base Sequence , Binding Sites , Humans , Protein Binding , Response Elements , Smad2 Protein/chemistry , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad4 Protein/genetics , Transcriptional Activation
16.
Cancer Sci ; 109(11): 3532-3542, 2018 Nov.
Article En | MEDLINE | ID: mdl-30207029

The PHLDA family (pleckstrin homology-like domain family) of genes consists of 3 members: PHLDA1, 2, and 3. Both PHLDA3 and PHLDA2 are phosphatidylinositol (PIP) binding proteins and function as repressors of Akt. They have tumor suppressive functions, mainly through Akt inhibition. Several reports suggest that PHLDA1 also has a tumor suppressive function; however, the precise molecular functions of PHLDA1 remain to be elucidated. Through a comprehensive screen for p53 target genes, we identified PHLDA1 as a novel p53 target, and we show that PHLDA1 has the ability to repress Akt in a manner similar to that of PHLDA3 and PHLDA2. PHLDA1 has a so-called split PH domain in which the PH domain is divided into an N-terminal (ß sheets 1-3) and a C-terminal (ß sheets 4-7 and an α-helix) portions. We show that the PH domain of PHLDA1 is responsible for its localization to the plasma membrane and binding to phosphatidylinositol. We also show that the function of the PH domain is essential for Akt repression. In addition, PHLDA1 expression analysis suggests that PHLDA1 has a tumor suppressive function in breast and ovarian cancers.


Breast Neoplasms/genetics , Ovarian Neoplasms/genetics , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Alternative Splicing , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Neoplasm Transplantation , Phosphatidylinositols/metabolism , Protein Binding , Transcription Factors/chemistry
17.
Cancer Sci ; 109(9): 2907-2918, 2018 Sep.
Article En | MEDLINE | ID: mdl-29945296

EWS-FLI1 constitutes an oncogenic transcription factor that plays key roles in Ewing sarcoma development and maintenance. We have recently succeeded in generating an ex vivo mouse model for Ewing sarcoma by introducing EWS-FLI1 into embryonic osteochondrogenic progenitors. The model well recapitulates the biological characteristics, small round cell morphology, and gene expression profiles of human Ewing sarcoma. Here, we clarified the global DNA binding properties of EWS-FLI1 in mouse Ewing sarcoma. GGAA microsatellites were found to serve as binding sites of EWS-FLI1 albeit with less frequency than that in human Ewing sarcoma; moreover, genomic distribution was not conserved between human and mouse. Nevertheless, EWS-FLI1 binding sites within GGAA microsatellites were frequently associated with the histone H3K27Ac enhancer mark, suggesting that EWS-FLI1 could affect global gene expression by binding its target sites. In particular, the Fox transcription factor binding motif was frequently observed within EWS-FLI1 peaks and Foxq1 was identified as the cooperative partner that interacts with the EWS portion of EWS-FLI1. Trib1 and Nrg1 were demonstrated as target genes that are co-regulated by EWS-FLI1 and Foxq1, and are important for cell proliferation and survival of Ewing sarcoma. Collectively, our findings present novel aspects of EWS-FLI1 function as well as the importance of GGAA microsatellites.


Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/genetics , Microsatellite Repeats/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/pathology , Animals , Apoptosis/genetics , Binding Sites/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Chondrogenesis/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Embryonic Stem Cells/cytology , Epithelial-Mesenchymal Transition/genetics , Forkhead Transcription Factors/genetics , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Neuregulin-1/genetics , Oncogene Proteins, Fusion/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Protein c-fli-1/genetics , RNA Interference , RNA, Small Interfering/genetics , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/genetics
18.
Nat Cell Biol ; 19(9): 1081-1092, 2017 Sep.
Article En | MEDLINE | ID: mdl-28812581

Brown fat dissipates energy as heat and protects against obesity. Here, we identified nuclear factor I-A (NFIA) as a transcriptional regulator of brown fat by a genome-wide open chromatin analysis of murine brown and white fat followed by motif analysis of brown-fat-specific open chromatin regions. NFIA and the master transcriptional regulator of adipogenesis, PPARγ, co-localize at the brown-fat-specific enhancers. Moreover, the binding of NFIA precedes and facilitates the binding of PPARγ, leading to increased chromatin accessibility and active transcription. Introduction of NFIA into myoblasts results in brown adipocyte differentiation. Conversely, the brown fat of NFIA-knockout mice displays impaired expression of the brown-fat-specific genes and reciprocal elevation of muscle genes. Finally, expression of NFIA and the brown-fat-specific genes is positively correlated in human brown fat. These results indicate that NFIA activates the cell-type-specific enhancers and facilitates the binding of PPARγ to control the brown fat gene program.


Adipocytes, Brown/metabolism , Adipogenesis/genetics , Adipose Tissue, Brown/metabolism , NFI Transcription Factors/metabolism , PPAR gamma/metabolism , Transcription, Genetic , 3T3-L1 Cells , Adipose Tissue, Brown/cytology , Animals , Binding Sites , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , Genotype , High-Throughput Nucleotide Sequencing , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Muscle Development , Myoblasts/metabolism , NFI Transcription Factors/deficiency , NFI Transcription Factors/genetics , PPAR gamma/genetics , Phenotype , Protein Binding , RNA Interference , Signal Transduction , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
19.
Sci Rep ; 7(1): 1166, 2017 04 26.
Article En | MEDLINE | ID: mdl-28446749

Epithelial-mesenchymal transition (EMT) is induced by transforming growth factor (TGF)-ß and facilitates tumor progression. We here performed global mapping of accessible chromatin in the mouse mammary gland epithelial EpH4 cell line and its Ras-transformed derivative (EpRas) using formaldehyde-assisted isolation of regulatory element (FAIRE)-sequencing. TGF-ß and Ras altered chromatin accessibility either cooperatively or independently, and AP1, ETS, and RUNX binding motifs were enriched in the accessible chromatin regions of EpH4 and EpRas cells. Etv4, an ETS family oncogenic transcription factor, was strongly expressed and bound to more than one-third of the accessible chromatin regions in EpRas cells treated with TGF-ß. While knockdown of Etv4 and another ETS family member Etv5 showed limited effects on the decrease in the E-cadherin abundance and stress fiber formation by TGF-ß, gene ontology analysis showed that genes encoding extracellular proteins were most strongly down-regulated by Etv4 and Etv5 siRNAs. Accordingly, TGF-ß-induced expression of Mmp13 and cell invasiveness were suppressed by Etv4 and Etv5 siRNAs, which were accompanied by the reduced chromatin accessibility at an enhancer region of Mmp13 gene. These findings suggest a mechanism of transcriptional regulation during Ras- and TGF-ß-induced EMT that involves alterations of accessible chromatin, which are partly regulated by Etv4 and Etv5.


Cell Transformation, Neoplastic , Chromatin/metabolism , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition , Mammary Glands, Animal/cytology , Proto-Oncogene Proteins p21(ras)/metabolism , Transforming Growth Factor beta/metabolism , Animals , Binding Sites , Cell Line , DNA/metabolism , Gene Expression Regulation , Mice , Protein Binding
20.
PLoS One ; 11(6): e0157992, 2016.
Article En | MEDLINE | ID: mdl-27336669

TGF-ß is a multifunctional cytokine that is involved in cell proliferation, differentiation and function. We previously reported an essential role of the TGF-ß -Smad2/3 pathways in RANKL-induced osteoclastogenesis. Using chromatin immunoprecipitation followed by sequencing, we comprehensively identified Smad2/3 target genes in bone marrow macrophages. These genes were enriched in the gene population upregulated by TGF-ß and downregulated by RANKL. Recent studies have revealed that histone modifications, such as trimethylation of histone H3 lysine 4 (H3K4me3) and lysine 27 (H3K27me3), critically regulate key developmental steps. We identified Nedd9 as a Smad2/3 target gene whose histone modification pattern was converted from H3K4me3(+)/H3K4me27(+) to H3K4me3(+)/H3K4me27(-) by TGF-ß. Nedd9 expression was increased by TGF-ß and suppressed by RANKL. Overexpression of Nedd9 partially rescued an inhibitory effect of a TGF-ß inhibitor, while gene silencing of Nedd9 suppressed RANKL-induced osteoclastogenesis. RANKL-induced osteoclastogenesis were reduced and stimulatory effects of TGF-ß on RANKL-induced osteoclastogenesis were partially abrogated in cells from Nedd9-deficient mice although knockout mice did not show abnormal skeletal phenotypes. These results suggest that Nedd9 is a Smad2/3 target gene implicated in RANKL-induced osteoclastogenesis.


Adaptor Proteins, Signal Transducing/genetics , Bone Resorption/genetics , Gene Expression Regulation , Osteoclasts/metabolism , RANK Ligand/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Bone Resorption/drug therapy , Cell Differentiation/drug effects , Cell Differentiation/genetics , Epigenesis, Genetic , Gene Expression Profiling , Histones/metabolism , Male , Mice , Mice, Knockout , Osteoclasts/drug effects , Protein Binding , RANK Ligand/pharmacology
...