Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Mol Cell ; 81(7): 1411-1424.e7, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33567268

ABSTRACT

Targeted protein degradation is an emerging therapeutic paradigm. Small-molecule degraders such as proteolysis-targeting chimeras (PROTACs) induce the degradation of neo-substrates by hijacking E3 ubiquitin ligases. Although ubiquitylation of endogenous substrates has been extensively studied, the mechanism underlying forced degradation of neo-substrates is less well understood. We found that the ubiquitin ligase TRIP12 promotes PROTAC-induced and CRL2VHL-mediated degradation of BRD4 but is dispensable for the degradation of the endogenous CRL2VHL substrate HIF-1α. TRIP12 associates with BRD4 via CRL2VHL and specifically assembles K29-linked ubiquitin chains, facilitating the formation of K29/K48-branched ubiquitin chains and accelerating the assembly of K48 linkage by CRL2VHL. Consequently, TRIP12 promotes the PROTAC-induced apoptotic response. TRIP12 also supports the efficiency of other degraders that target CRABP2 or TRIM24 or recruit CRBN. These observations define TRIP12 and K29/K48-branched ubiquitin chains as accelerators of PROTAC-directed targeted protein degradation, revealing a cooperative mechanism of branched ubiquitin chain assembly unique to the degradation of neo-substrates.


Subject(s)
Carrier Proteins/metabolism , Polyubiquitin/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , HCT116 Cells , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Polyubiquitin/genetics , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics
2.
Genes Dev ; 35(21-22): 1431-1444, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34675062

ABSTRACT

During neocortical development, tight regulation of neurogenesis-to-astrogenesis switching of neural precursor cells (NPCs) is critical to generate a balanced number of each neural cell type for proper brain functions. Accumulating evidence indicates that a complex array of epigenetic modifications and the availability of extracellular factors control the timing of neuronal and astrocytic differentiation. However, our understanding of NPC fate regulation is still far from complete. Bone morphogenetic proteins (BMPs) are renowned as cytokines that induce astrogenesis of gliogenic late-gestational NPCs. They also promote neurogenesis of mid-gestational NPCs, although the underlying mechanisms remain elusive. By performing multiple genome-wide analyses, we demonstrate that Smads, transcription factors that act downstream from BMP signaling, target dramatically different genomic regions in neurogenic and gliogenic NPCs. We found that histone H3K27 trimethylation and DNA methylation around Smad-binding sites change rapidly as gestation proceeds, strongly associated with the alteration of accessibility of Smads to their target binding sites. Furthermore, we identified two lineage-specific Smad-interacting partners-Sox11 for neurogenic and Sox8 for astrocytic differentiation-that further ensure Smad-regulated fate-specific gene induction. Our findings illuminate an exquisite regulation of NPC property change mediated by the interplay between cell-extrinsic cues and -intrinsic epigenetic programs during cortical development.


Subject(s)
Neural Stem Cells , Brain , Cell Differentiation/genetics , Epigenesis, Genetic , Female , Genome-Wide Association Study , Humans , Neurogenesis/genetics , Pregnancy , SOXE Transcription Factors/genetics
3.
Development ; 148(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34519339

ABSTRACT

Notch-Delta signaling regulates many developmental processes, including tissue homeostasis and maintenance of stem cells. Upon interaction of juxtaposed cells via Notch and Delta proteins, intracellular domains of both transmembrane proteins are cleaved and translocate to the nucleus. Notch intracellular domain activates target gene expression; however, the role of the Delta intracellular domain remains elusive. Here, we show the biological function of Delta like 1 intracellular domain (D1ICD) by modulating its production. We find that the sustained production of D1ICD abrogates cell proliferation but enhances neurogenesis in the developing dorsal root ganglia (DRG), whereas inhibition of D1ICD production promotes cell proliferation and gliogenesis. D1ICD acts as an integral component of lateral inhibition mechanism by inhibiting Notch activity. In addition, D1ICD promotes neurogenesis in a Notch signaling-independent manner. We show that D1ICD binds to Erk1/2 in neural crest stem cells and inhibits the phosphorylation of Erk1/2. In summary, our results indicate that D1ICD regulates DRG development by modulating not only Notch signaling but also the MAP kinase pathway.


Subject(s)
Calcium-Binding Proteins/metabolism , MAP Kinase Signaling System , Neurogenesis , Receptors, Notch/metabolism , Animals , Binding Sites , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Cell Proliferation , Cells, Cultured , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NIH 3T3 Cells , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Protein Binding
4.
Development ; 145(1)2018 01 08.
Article in English | MEDLINE | ID: mdl-29180567

ABSTRACT

Transcriptional regulation can be tightly orchestrated by epigenetic regulators. Among these, ubiquitin-like with PHD and RING finger domains 1 (Uhrf1) is reported to have diverse epigenetic functions, including regulation of DNA methylation. However, the physiological functions of Uhrf1 in skeletal tissues remain unclear. Here, we show that limb mesenchymal cell-specific Uhrf1 conditional knockout mice (Uhrf1ΔLimb/ΔLimb ) exhibit remarkably shortened long bones that have morphological deformities due to dysregulated chondrocyte differentiation and proliferation. RNA-seq performed on primary cultured chondrocytes obtained from Uhrf1ΔLimb/ΔLimb mice showed abnormal chondrocyte differentiation. In addition, integrative analyses using RNA-seq and MBD-seq revealed that Uhrf1 deficiency decreased genome-wide DNA methylation and increased gene expression through reduced DNA methylation in the promoter regions of 28 genes, including Hspb1, which is reported to be an IL1-related gene and to affect chondrocyte differentiation. Hspb1 knockdown in cKO chondrocytes can normalize abnormal expression of genes involved in chondrocyte differentiation, such as Mmp13 These results indicate that Uhrf1 governs cell type-specific transcriptional regulation by controlling the genome-wide DNA methylation status and regulating consequent cell differentiation and skeletal maturation.


Subject(s)
Cell Differentiation/physiology , Chondrocytes/metabolism , Gene Expression Regulation/physiology , Hindlimb/growth & development , Musculoskeletal Development/physiology , Nuclear Proteins/metabolism , Animals , CCAAT-Enhancer-Binding Proteins , DNA Methylation/physiology , Genome-Wide Association Study , Mice , Mice, Knockout , Nuclear Proteins/genetics , Promoter Regions, Genetic/physiology , Ubiquitin-Protein Ligases
5.
Brain ; 142(6): 1675-1689, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31135049

ABSTRACT

The mechanism by which dopaminergic neurons are selectively affected in Parkinson's disease is not fully understood. In this study, we found a dramatic increase in the expression of catechol-O-methyltransferase (COMT), along with a lower level of DNA methylation, in induced pluripotent stem cell-derived dopaminergic neurons from patients with parkin (PARK2) gene mutations compared to those from healthy controls. In addition, a significant increase in the expression of COMT was found in dopaminergic neurons of isogenic PARK2 induced pluripotent stem cell lines that mimicked loss of function of PARK2 by CRISPR Cas9 technology. In dopamine transporter (DAT)-Cre mice, overexpression of COMT, specifically in dopaminergic neurons of the substantia nigra, produced cataleptic behaviours associated with impaired motor coordination. These findings suggest that upregulation of COMT, likely resulting from DNA hypomethylation, in dopaminergic neurons may contribute to the initial stage of neuronal dysfunction in Parkinson's disease.


Subject(s)
Catechol O-Methyltransferase/genetics , Dopaminergic Neurons/metabolism , Parkinson Disease/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice, Transgenic , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Substantia Nigra/metabolism
6.
Mol Pharmacol ; 96(5): 600-608, 2019 11.
Article in English | MEDLINE | ID: mdl-31455676

ABSTRACT

Induction of cytochrome P450 enzyme 3A (CYP3A) in response to pregnane X receptor (PXR) activators shows species-specific differences. To study the induction of human CYP3A in response to human PXR activators, we generated a double-humanized mouse model of PXR and CYP3A. CYP3A-humanized mice generated by using a mouse artificial chromosome (MAC) vector containing the entire genomic human CYP3A locus (hCYP3A-MAC mouse line) were bred with PXR-humanized mice in which the ligand-binding domain of mouse PXR was replaced with that of human PXR, resulting in double-humanized mice (hCYP3A-MAC/hPXR mouse line). Oral administration of the human PXR activator rifampicin increased hepatic expression of CYP3A4 mRNA and triazolam (TRZ) 1'- and 4-hydroxylation activities, CYP3A probe activities, in the liver and intestine microsomes of hCYP3A-MAC/hPXR mice. The plasma concentration of TRZ after oral dosing was significantly decreased by rifampicin treatment in hCYP3A-MAC/hPXR mice but not in hCYP3A-MAC mice. In addition, mass spectrometry imaging analysis showed that rifampicin treatment increased the formation of hydroxy TRZ in the intestine of hCYP3A-MAC/hPXR mice after oral dosing of TRZ. The plasma concentration of 1'- and 4-hydroxy TRZ in portal blood was also increased by rifampicin treatment in hCYP3A-MAC/hPXR mice. These results suggest that the hCYP3A-MAC/hPXR mouse line may be a useful model to predict human PXR-dependent induction of metabolism of CYP3A4 substrates in the liver and intestine. SIGNIFICANCE STATEMENT: We generated a double-humanized mouse line for CYP3A and PXR. Briefly, CYP3A-humanized mice generated by using a mouse artificial chromosome vector containing the entire genomic human CYP3A locus were bred with PXR-humanized mice in which the ligand-binding domain of mouse PXR was replaced with that of human PXR. Expression of CYP3A4 and metabolism of triazolam, a typical CYP3A substrate, in the liver of CYP3A/PXR-humanized mice were enhanced in response to rifampicin, a typical human PXR activator. Enhancement of triazolam metabolism in the intestine of CYP3A/PXR-humanized mice was firstly shown by combination of mass spectrometry imaging of sliced intestine and liquid chromatography with tandem mass spectrometry analysis of metabolite concentration in portal blood after oral dosing of triazolam.


Subject(s)
Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A/biosynthesis , Intestine, Small/metabolism , Liver/metabolism , Portal Vein/metabolism , Pregnane X Receptor/biosynthesis , Animals , Enzyme Induction/drug effects , Enzyme Induction/physiology , Humans , Intestine, Small/drug effects , Liver/drug effects , Mass Spectrometry/methods , Mice , Mice, Knockout , Mice, Transgenic , Portal Vein/drug effects
7.
Biochem Biophys Res Commun ; 498(3): 674-679, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29524425

ABSTRACT

Testicular toxicity is a frequent adverse effect of cancer chemotherapy that has no effective clinical biomarker. To find new biomarkers, we focused on epigenetic mechanisms in the male germline. We investigated the DNA methylation status of the male germline during testicular toxicity induced by doxorubicin (DXR), a widely used anticancer agent. We established mouse models of early stage testicular toxicity and testicular pre-toxicity by the administration of 0.2 mg/kg and 0.02 mg/kg DXR, respectively, twice weekly for 5 weeks. Histological analysis showed sparse abnormalities in testicular tissue; however, western blotting analysis revealed reduced testicular expression levels of DNA methyltransferases DNMT3a and DNMT3b in both DXR-treated groups. Interestingly, comprehensive sperm DNA methylation analysis using Methyl-CpG binding domain protein-enriched genome sequencing revealed that hypomethylation was the most frequent change induced by DXR. These findings suggest that sperm DNA methylation status may be used as an early diagnostic marker for testicular changes not detected by conventional toxicity analysis.


Subject(s)
Antibiotics, Antineoplastic/toxicity , DNA Methylation/drug effects , Doxorubicin/toxicity , Spermatozoa/drug effects , Testis/drug effects , Animals , DNA (Cytosine-5-)-Methyltransferases/analysis , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , Male , Mice , Mice, Inbred C57BL , Sperm Motility/drug effects , Spermatozoa/metabolism , Spermatozoa/pathology , Testis/metabolism , Testis/pathology , DNA Methyltransferase 3B
8.
Development ; 141(11): 2260-70, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24821986

ABSTRACT

Retinoic acid receptor gamma 2 (RARγ2) is the major RAR isoform expressed throughout the caudal axial progenitor domain in vertebrates. During a microarray screen to identify RAR targets, we identified a subset of genes that pattern caudal structures or promote axial elongation and are upregulated by increased RAR-mediated repression. Previous studies have suggested that RAR is present in the caudal domain, but is quiescent until its activation in late stage embryos terminates axial elongation. By contrast, we show here that RARγ2 is engaged in all stages of axial elongation, not solely as a terminator of axial growth. In the absence of RA, RARγ2 represses transcriptional activity in vivo and maintains the pool of caudal progenitor cells and presomitic mesoderm. In the presence of RA, RARγ2 serves as an activator, facilitating somite differentiation. Treatment with an RARγ-selective inverse agonist (NRX205099) or overexpression of dominant-negative RARγ increases the expression of posterior Hox genes and that of marker genes for presomitic mesoderm and the chordoneural hinge. Conversely, when RAR-mediated repression is reduced by overexpressing a dominant-negative co-repressor (c-SMRT), a constitutively active RAR (VP16-RARγ2), or by treatment with an RARγ-selective agonist (NRX204647), expression of caudal genes is diminished and extension of the body axis is prematurely terminated. Hence, gene repression mediated by the unliganded RARγ2-co-repressor complex constitutes a novel mechanism to regulate and facilitate the correct expression levels and spatial restriction of key genes that maintain the caudal progenitor pool during axial elongation in Xenopus embryos.


Subject(s)
Gene Expression Regulation, Developmental , Receptors, Retinoic Acid/metabolism , Animals , Apoptosis , Cell Differentiation/genetics , Co-Repressor Proteins/metabolism , Gene Expression Regulation , Genes, Dominant , Homeodomain Proteins/metabolism , Humans , Mesoderm/metabolism , Mesoderm/physiology , Nervous System/embryology , Nervous System/growth & development , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Receptors, Retinoic Acid/agonists , Repressor Proteins/metabolism , Retinoic Acid Receptor alpha , Signal Transduction , Somites/physiology , Time Factors , Xenopus Proteins/metabolism , Xenopus laevis , Retinoic Acid Receptor gamma
9.
Breast Cancer Res Treat ; 161(2): 269-278, 2017 01.
Article in English | MEDLINE | ID: mdl-27888420

ABSTRACT

PURPOSE: Triple-negative breast cancer (TNBC) has aggressive characteristics and fewer treatment options than other subtypes. The purpose of this study was to explore prognostic biomarkers for TNBC that can be easily detected from the blood samples. METHODS: MDA-MB-231 and MDA-MB-231BR, a brain metastatic variant of the human TNBC cell line MDA-MB-231, were used as less and more aggressive models of TNBC, respectively. The extent to which the candidate gene/protein identified by RNA sequencing correlated well with aggressiveness of TNBC and how much protein was detected from the blood of tumor-bearing mice were evaluated. RESULTS: Both the in vitro proliferation and in vivo tumor growth of MDA-MB-231BR were more rapid than those of MDA-MB-231. RNA sequencing identified ESM1 as a gene that was expressed significantly more in MDA-MB-231BR than in MDA-MB-231, and qRT-PCR confirmed a significantly higher expression of ESM1 in MDA-MB-231BR xenograft in vivo. Furthermore, Kaplan-Meier analysis of relapse-free survival demonstrated that TNBC patients with high ESM1 expression had clearly worse relapse-free survival than those with low ESM1 expression, which was consistent with our preclinical findings. Endocan, a protein of ESM1 gene product, was successfully detected in both conditioned medium from MDA-MB-231BR and plasma samples from mice bearing MDA-MB-231BR xenograft, which showed a significantly distinct pattern from less aggressive MDA-MB-231. Moreover, bisulfite sequence analysis revealed that overexpression of ESM1 in MDA-MB-231BR might be attributed to DNA demethylation in an upstream region of the ESM1 gene. CONCLUSION: This study indicates that endocan could be used as a blood-based prognostic biomarker in TNBC patients.


Subject(s)
Biomarkers, Tumor , Neoplasm Proteins/metabolism , Proteoglycans/metabolism , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/mortality , Animals , Cell Line, Tumor , CpG Islands , DNA Methylation , Disease Models, Animal , Extracellular Space/metabolism , Female , Gene Expression , Heterografts , Humans , Mice , Neoplasm Proteins/blood , Neoplasm Proteins/genetics , Prognosis , Proteoglycans/blood , Proteoglycans/genetics , Triple Negative Breast Neoplasms/genetics
10.
Nature ; 480(7378): 557-60, 2011 Nov 27.
Article in English | MEDLINE | ID: mdl-22121020

ABSTRACT

Chromatin reorganization is governed by multiple post-translational modifications of chromosomal proteins and DNA. These histone modifications are reversible, dynamic events that can regulate DNA-driven cellular processes. However, the molecular mechanisms that coordinate histone modification patterns remain largely unknown. In metazoans, reversible protein modification by O-linked N-acetylglucosamine (GlcNAc) is catalysed by two enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). However, the significance of GlcNAcylation in chromatin reorganization remains elusive. Here we report that histone H2B is GlcNAcylated at residue S112 by OGT in vitro and in living cells. Histone GlcNAcylation fluctuated in response to extracellular glucose through the hexosamine biosynthesis pathway (HBP). H2B S112 GlcNAcylation promotes K120 monoubiquitination, in which the GlcNAc moiety can serve as an anchor for a histone H2B ubiquitin ligase. H2B S112 GlcNAc was localized to euchromatic areas on fly polytene chromosomes. In a genome-wide analysis, H2B S112 GlcNAcylation sites were observed widely distributed over chromosomes including transcribed gene loci, with some sites co-localizing with H2B K120 monoubiquitination. These findings suggest that H2B S112 GlcNAcylation is a histone modification that facilitates H2BK120 monoubiquitination, presumably for transcriptional activation.


Subject(s)
Acetylglucosamine/metabolism , Histones/metabolism , Amino Acid Sequence , Animals , Cell Line , HeLa Cells , Histones/chemistry , Histones/genetics , Humans , Models, Molecular , Mutation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Ubiquitination
11.
Biol Pharm Bull ; 40(1): 113-117, 2017.
Article in English | MEDLINE | ID: mdl-28049944

ABSTRACT

Mesenchymal stem cells (MSCs) have been explored as a "live" carrier of cytokines for targeted cancer therapy, but, in earlier reports in the literature, the secretion process of therapeutic cytokines was not regulated. The purpose of this study was to generate MSCs to conditionally secrete the melanoma differentiation-associated gene-7 (MDA-7) tumor-suppressor protein. To control the secretion of MDA-7 from MSCs, a well-established tetracycline-controlled transcriptional activation system was incorporated into MDA-7 plasmid. MDA-7 gene expression was induced in the engineered MSCs only in the presence of doxycycline, as characterized by quantitative reverse transcription (qRT)-PCR. Enzyme-linked immunosorbent assay (ELISA) also revealed that the MDA-7 protein was secreted from the engineered MSCs only after the cells had been exposed to doxycycline. Both recombinant human MDA-7 protein and the conditioned medium from the engineered MSCs in the presence of doxycycline significantly inhibited tube formation of human umbilical vascular endothelial cells (HUVECs), indicating that our system could be used for targeted, antiangiogenic therapy. Overall, this study provides useful information on the potential use of engineered MSCs for the controlled secretion of therapeutic proteins, in this case MDA-7, for targeted cancer therapy.


Subject(s)
Doxycycline/pharmacology , Interleukins/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Neoplasms/therapy , Animals , Gene Expression , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Interleukins/genetics , Mesenchymal Stem Cells/drug effects , Mice, Inbred BALB C , Neoplasms/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
Synapse ; 70(8): 317-24, 2016 08.
Article in English | MEDLINE | ID: mdl-26990296

ABSTRACT

A multiplex analysis for profiling the expression of candidate microRNAs (miRNAs), which are small noncoding RNAs that function as key post-transcriptional regulators, may lead to a better understanding of the complex machinery of neuropathic pain. In the present study, we performed a miRNA array analysis using tissues of the dorsal root ganglion (DRG), a primary site for pain processing, obtained from mice with partial sciatic nerve ligation. Among 1135 total miRNAs, 26 miRNAs showed up-regulation (more than 2-fold change) and only 4 miRNAs showed down-regulation (less than 0.5-fold change) in the DRG of nerve-ligated mice. In a RT-qPCR assay, the levels of miR-21, miR-431, and miR-511-3p were significantly increased on the ipsilateral side of the DRG from 3 to 7 days after sciatic nerve ligation. These elevations were almost absent in IL-6 knockout mice. Furthermore, the expression level of miR-21, but not those of miR-431 or miR511-3p, was significantly increased in exosomes extracted from blood of nerve-ligated mice. These findings suggest that the increased expression of IL-6-regulated miR-21, miR-431, and miR-511-3p in the DRG and increased exosomal miR-21 extracted from blood after sciatic nerve ligation may play at least a partial role in neuropathic pain. Synapse 70:317-324, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Ganglia, Spinal/metabolism , Interleukin-6/metabolism , MicroRNAs/genetics , Neuralgia/metabolism , Animals , Exosomes/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Neuralgia/genetics , Sensory Receptor Cells/metabolism
13.
J Biol Chem ; 289(26): 18152-62, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24828497

ABSTRACT

Regulation of spatiotemporal gene expression in higher eukaryotic cells is critical for the precise and orderly development of undifferentiated progenitors into committed cell types of the adult. It is well known that dynamic epigenomic regulation (including chromatin remodeling and histone modifications by transcriptional coregulator complexes) is involved in transcriptional regulation. Precisely how these coregulator complexes exert their cell type and developing stage-specific activity is largely unknown. In this study we aimed to isolate the histone demethylase lysine-specific demethylase 1 (LSD1) complex from neural cells by biochemical purification. In so doing, we identified myelin transcription factor 1 (MyT1) as a novel LSD1 complex component. MyT1 is a neural cell-specific zinc finger factor, and it forms a stable multiprotein complex with LSD1 through direct interaction. Target gene analysis using microarray and ChIP assays revealed that the Pten gene was directly regulated by the LSD1-MyT1 complex. Knockdown of either LSD1 or MyT1 derepressed the expression of endogenous target genes and inhibited cell proliferation of a neuroblastoma cell line, Neuro2a. We propose that formation of tissue-specific combinations of coregulator complexes is a critical mechanism for tissue-specific transcriptional regulation.


Subject(s)
DNA-Binding Proteins/metabolism , Neurons/enzymology , Oxidoreductases, N-Demethylating/metabolism , Transcription Factors/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , Histone Demethylases , Histones/metabolism , Mice , Neurons/metabolism , Oxidoreductases, N-Demethylating/genetics , Protein Subunits/genetics , Protein Subunits/metabolism , Species Specificity , Transcription Factors/genetics
14.
Article in Japanese | MEDLINE | ID: mdl-26434097

ABSTRACT

Genetic factors are involved in determinants for the risk of psychiatric disorders, and neurological and neurodegenerative diseases. Chronic pain stimuli and intense pain have effects at a cellular and/or gene expression level, and will eventually induce "cellular memory due to pain", which means that tissue damage, even if only transient, can elicit epigenetically abnormal transcription/translation and post-translational modification in related cells depending on the degree or kind of injury or associated conditions. Such cell memory/transformation due to pain can cause an abnormality in a fundamental intracellular response, such as a change in the three-dimensional structure of DNA, transcription, or translation. On the other hand, pain is a multidimensional experience with sensory-discriminative and motivational-affective components. Recent human brain imaging studies have examined differences in activity in the nucleus accumbens between controls and patients with chronic pain, and have revealed that the nucleus accumbens plays a role in predicting the value of a noxious stimulus and its offset, and in the consequent changes in the motivational state. In this review, we provide a very brief overview of a comprehensive understanding of chronic pain associated with emotional dysregulation due to transcriptional regulation, epigenetic modification and miRNA regulation.


Subject(s)
Pain/physiopathology , Dopamine/physiology , Emotions/physiology , Epigenomics , Humans
15.
Biochem Biophys Res Commun ; 437(4): 579-84, 2013 Aug 09.
Article in English | MEDLINE | ID: mdl-23850677

ABSTRACT

Chondrocyte differentiation is controlled by various regulators, such as Sox9 and Runx2, but the process is complex. To further understand the precise underlying molecular mechanisms of chondrocyte differentiation, we aimed to identify a novel regulatory factor of chondrocyte differentiation using gene expression profiles of micromass-cultured chondrocytes at different differentiation stages. From the results of microarray analysis, the autoimmune regulator, Aire, was identified as a novel regulator. Aire stable knockdown cells, and primary cultured chondrocytes obtained from Aire(-/-) mice, showed reduced mRNA expression levels of chondrocyte-related genes. Over-expression of Aire induced the early stages of chondrocyte differentiation by facilitating expression of Bmp2. A ChIP assay revealed that Aire was recruited on an Airebinding site (T box) in the Bmp2 promoter region in the early stages of chondrocyte differentiation and histone methylation was modified. These results suggest that Aire can facilitate early chondrocyte differentiation by expression of Bmp2 through altering the histone modification status of the promoter region of Bmp2. Taken together, Aire might play a role as an active regulator of chondrocyte differentiation, which leads to new insights into the regulatory mechanisms of chondrocyte differentiation.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Cell Differentiation , Chondrocytes/cytology , Transcription Factors/metabolism , Animals , Chondrogenesis/genetics , Gene Expression Profiling , Gene Expression Regulation , Histones/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , AIRE Protein
16.
Proc Natl Acad Sci U S A ; 107(8): 3594-9, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20133598

ABSTRACT

Nanos is one of the evolutionarily conserved proteins implicated in germ cell development. We have previously shown that NANOS2 plays an important role in both the maintenance and sexual development of germ cells. However, the molecular mechanisms underlying these events have remained elusive. In our present study, we found that NANOS2 localizes to the P-bodies, known centers of RNA degradation that are abundantly accumulated in male gonocytes. We further identified by immunoprecipitation that the components of the CCR4-NOT deadenylation complex are NANOS2-interacting proteins and found that NANOS2 promotes the localization of CNOT proteins to P-bodies in vivo. We also elucidated that the NANOS2/CCR4-NOT complex has deadenylase activity in vitro, and that some of the RNAs implicated in meiosis interact with NANOS2 and are accumulated in its absence. Our current data thus indicate that the expression of these RNA molecules is normally suppressed via a NANOS2-mediated mechanism. We propose from our current findings that NANOS2-interacting RNAs may be recruited to P-bodies and degraded by the enzymes contained therein through NANOS2-mediated deadenylation.


Subject(s)
Carrier Proteins/metabolism , RNA Stability , Ribonucleases/metabolism , Spermatozoa/enzymology , Transcription Factors/metabolism , Animals , Carrier Proteins/genetics , Male , Mice , Mice, Knockout , RNA, Messenger/metabolism , RNA-Binding Proteins , Ribonucleases/genetics
17.
J Clin Invest ; 132(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35472067

ABSTRACT

Rheumatoid arthritis (RA) is characterized by chronic synovial inflammation with aberrant epigenetic alterations, eventually leading to joint destruction. However, the epigenetic regulatory mechanisms underlying RA pathogenesis remain largely unknown. Here, we showed that ubiquitin-like containing PHD and RING finger domains 1 (UHRF1) is a central epigenetic regulator that orchestrates multiple pathogeneses in RA in a suppressive manner. UHRF1 expression was remarkably upregulated in synovial fibroblasts (SFs) from arthritis model mice and patients with RA. Mice with SF-specific Uhrf1 conditional knockout showed more severe arthritic phenotypes than littermate controls. Uhrf1-deficient SFs also exhibited enhanced apoptosis resistance and upregulated expression of several cytokines, including Ccl20. In patients with RA, DAS28, CRP, and Th17 accumulation and apoptosis resistance were negatively correlated with UHRF1 expression in synovium. Finally, Ryuvidine administration stabilized UHRF1 ameliorated arthritis pathogeneses in a mouse model of RA. This study demonstrated that UHRF1 expressed in RA SFs can contribute to negative feedback mechanisms that suppress multiple pathogenic events in arthritis, suggesting that targeting UHRF1 could be one of the therapeutic strategies for RA.


Subject(s)
Arthritis, Rheumatoid , CCAAT-Enhancer-Binding Proteins , Epigenesis, Genetic , Ubiquitin-Protein Ligases , Animals , Arthritis, Rheumatoid/pathology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Fibroblasts/metabolism , Gene Expression , Humans , Inflammation/pathology , Mice , Synovial Membrane/pathology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
18.
Hippocampus ; 21(2): 127-32, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20232397

ABSTRACT

Environmental enrichment is an experimental paradigm that increases brain-derived neurotrophic factor (BDNF) gene expression accompanied by neurogenesis in the hippocampus of rodents. In the present study, we investigated whether an enriched environment could cause epigenetic modification at the BDNF gene in the hippocampus of mice. Exposure to an enriched environment for 3-4 weeks caused a dramatic increase in the mRNA expression of BDNF, but not platelet-derived growth factor A (PDGF-A), PDGF-B, vascular endothelial growth factor (VEGF), nerve growth factor (NGF), epidermal growth factor (EGF), or glial fibrillary acidic protein (GFAP), in the hippocampus of mice. Under these conditions, exposure to an enriched environment induced a significant increase in histone H3 lysine 4 (H3K4) trimethylation at the BDNF P3 and P6 promoters, in contrast to significant decreases in histone H3 lysine 9 (H3K9) trimethylation at the BDNF P4 promoter and histone H3 lysine 27 (H3K27) trimethylation at the BDNF P3 and P4 promoters without any changes in the expression of their associated histone methylases and demethylases in the hippocampus. The expression levels of several microRNAs in the hippocampus were not changed by an enriched environment. These results suggest that an enriched environment increases BDNF mRNA expression via sustained epigenetic modification in the mouse hippocampus.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Epigenesis, Genetic , Hippocampus/metabolism , Animals , Environment , Epidermal Growth Factor/genetics , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein , Hippocampus/growth & development , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factor/genetics , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Platelet-Derived Growth Factor/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-sis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/genetics
19.
Biochem Biophys Res Commun ; 412(2): 366-72, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21821006

ABSTRACT

It has recently been demonstrated that genistein (GEN), a phytoestrogen in soy products, is an epigenetic modulator in various types of cells; but its effect on endometrium has not yet been determined. We investigated the effects of GEN on mouse uterine cells, in vivo and in vitro. Oral administration of GEN for 1 week induced mild proliferation of the endometrium in ovariectomized (OVX) mice, which was accompanied by the induction of steroidogenic factor 1 (SF-1) gene expression. GEN administration induced demethylation of multiple CpG sites in the SF-1 promoter; these sites are extensively methylated and thus silenced in normal endometrium. The GEN-mediated promoter demethylation occurred predominantly on the luminal side, as opposed to myometrium side, indicating that the epigenetic change was mainly shown in regenerated cells. Primary cultures of endometrial stromal cell colonies were screened for GEN-mediated alterations of DNA methylation by a high-resolution melting (HRM) method. One out of 20 colony-forming cell clones showed GEN-induced demethylation of SF-1. This clone exhibited a high proliferation capacity with continuous colony formation activity through multiple serial clonings. We propose that only a portion of endometrial cells are capable of receiving epigenetic modulation by GEN.


Subject(s)
DNA Methylation/drug effects , Endometrium/drug effects , Epigenesis, Genetic/drug effects , Genistein/pharmacology , Steroidogenic Factor 1/genetics , Animals , Cells, Cultured , Endometrium/cytology , Endometrium/metabolism , Female , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism
20.
Biol Reprod ; 84(4): 734-42, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21123812

ABSTRACT

The balance between androgens and estrogens is very important in the development of the prostate, and even small changes in estrogen levels, including those of estrogen-mimicking chemicals, can lead to serious changes. Bisphenol A (BPA), an endocrine-disrupting chemical, is a well-known, ubiquitous, estrogenic chemical. To investigate the effects of fetal exposure to low-dose BPA on the development of the prostate, we examined alterations of the in situ sex steroid hormonal environment in the mouse urogenital sinus (UGS). In the BPA-treated UGS, estradiol (E(2)) levels and CYP19A1 (cytochrome P450 aromatase) activity were significantly increased compared with those of the untreated and diethylstilbestrol (DES)-treated UGS. The mRNAs of steroidogenic enzymes, Cyp19a1 and Cyp11a1, and the sex-determining gene, Nr5a1, were up-regulated specifically in the BPA-treated group. The up-regulation of mRNAs was observed in the mesenchymal component of the UGS as well as in the cerebellum, heart, kidney, and ovary but not in the testis. The number of aromatase-expressing mesenchymal cells in the BPA-treated UGS was approximately twice that in the untreated and DES-treated UGS. The up-regulation of Esrrg mRNA was observed in organs for which mRNAs of steroidogenic enzymes were also up-regulated. We demonstrate here that fetal exposure to low-dose BPA has the unique action of increasing in situ E(2) levels and CYP19A1 (aromatase) activity in the mouse UGS. Our data suggest that BPA might interact with in situ steroidogenesis by altering tissue components, such as the accumulation of aromatase-expressing mesenchymal cells, in particular organs.


Subject(s)
Endocrine Disruptors/toxicity , Estradiol/biosynthesis , Phenols/toxicity , Urogenital System/drug effects , Urogenital System/metabolism , Animals , Aromatase/genetics , Aromatase/metabolism , Base Sequence , Benzhydryl Compounds , DNA Primers/genetics , Female , Male , Mice , Mice, Inbred C57BL , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/genetics , Tissue Culture Techniques , Up-Regulation/drug effects , Urogenital System/embryology
SELECTION OF CITATIONS
SEARCH DETAIL