Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Life Sci ; 292: 120321, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35031259

ABSTRACT

Triple-negative breast cancer (TNBC) is a particularly lethal subtype of breast cancer owing to its heterogeneity, high drug resistance, poor prognosis and lack of therapeutic targets. Recent insights into the complexity of TNBC have been explained by epigenetic regulation and its ability to modulate certain oncogenes and tumour suppressor genes. This has opened an emerging area in anti-cancer therapy using epigenetic modulating drugs, highlighting the epigenetic reprogramming during tumorigenesis and tumour development. Histone methylation and demethylation are such dynamic epigenetic mechanisms mediated by histone methyltransferases (HMTs) and histone demethylases (HDMs), respectively. The interplay between HMTs and HDMs in histone methylation extrapolates their viability as druggable epigenetic targets in TNBC. In this review, we aim to summarize recent progress in the field of epigenetics focusing on HMTs and HDMs in TNBC development and their potential use in targeted therapy for TNBC management.


Subject(s)
Histone Demethylases/physiology , Histone Methyltransferases/physiology , Triple Negative Breast Neoplasms/metabolism , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Oncogenes
2.
DNA Cell Biol ; 40(9): 1177-1184, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34432529

ABSTRACT

Previous studies have shown that lysine-specific demethylase 1 (LSD1) could regulate cell cycle progression through demethylation. The 3'domain of HOX transcript antisense RNA (Hotair) combined with the LSD1/CoREST/REST complex helps LSD1 target the corresponding gene. However, its role in mice's myocardial regeneration is still unclear. The heart from neonatal mice shows strong myocardial regeneration ability, but this ability disappears 7 days after birth. Our study shows that the myocardial tissue highly expresses Hotair and Lsd1 within 1 week after birth, consistent with the myocardial regeneration time window. Knockdown Lsd1 or Hotair expression by RNA interference could inhibit myocardial regeneration and cardiomyocyte proliferation. Our results suggest that Hotair-mediated demethylation of LSD1 may play an important role in myocardial regeneration in neonatal mice.


Subject(s)
Histone Demethylases/physiology , Myocytes, Cardiac , RNA, Long Noncoding/physiology , Animals , Animals, Newborn , Cell Proliferation , Cells, Cultured , Female , Male , Methylation , Mice , Mice, Inbred ICR , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Regeneration
3.
PLoS Genet ; 17(7): e1009715, 2021 07.
Article in English | MEDLINE | ID: mdl-34329293

ABSTRACT

Histone methylation is dynamically regulated to shape the epigenome and adjust central nuclear processes including transcription, cell cycle control and DNA repair. Lysine-specific histone demethylase 2 (LSD2) has been implicated in multiple types of human cancers. However, its functions remain poorly understood. This study investigated the histone demethylase LSD2 homolog AMX-1 in C. elegans and uncovered a potential link between H3K4me2 modulation and DNA interstrand crosslink (ICL) repair. AMX-1 is a histone demethylase and mainly localizes to embryonic cells, the mitotic gut and sheath cells. Lack of AMX-1 expression resulted in embryonic lethality, a decreased brood size and disorganized premeiotic tip germline nuclei. Expression of AMX-1 and of the histone H3K4 demethylase SPR-5 is reciprocally up-regulated upon lack of each other and the mutants show increased H3K4me2 levels in the germline, indicating that AMX-1 and SPR-5 regulate H3K4me2 demethylation. Loss of AMX-1 function activates the CHK-1 kinase acting downstream of ATR and leads to the accumulation of RAD-51 foci and increased DNA damage-dependent apoptosis in the germline. AMX-1 is required for the proper expression of mismatch repair component MutL/MLH-1 and sensitivity against ICLs. Interestingly, formation of ICLs lead to ubiquitination-dependent subcellular relocalization of AMX-1. Taken together, our data suggest that AMX-1 functions in ICL repair in the germline.


Subject(s)
DNA Repair/genetics , Histone Demethylases/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Caenorhabditis elegans Proteins , Cell Nucleus/metabolism , DNA Damage/genetics , DNA Repair/physiology , Germ Cells/metabolism , Histone Demethylases/physiology , Histones/genetics , Methylation , Protein Processing, Post-Translational/genetics , Ubiquitination
4.
Sci Rep ; 11(1): 884, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441614

ABSTRACT

Trophoblast stem cell (TSC) is crucial to the formation of placenta in mammals. Histone demethylase JMJD2 (also known as KDM4) family proteins have been previously shown to support self-renewal and differentiation of stem cells. However, their roles in the context of the trophoblast lineage remain unclear. Here, we find that knockdown of Jmjd2b resulted in differentiation of TSCs, suggesting an indispensable role of JMJD2B/KDM4B in maintaining the stemness. Through the integration of transcriptome and ChIP-seq profiling data, we show that JMJD2B is associated with a loss of H3K36me3 in a subset of embryonic lineage genes which are marked by H3K9me3 for stable repression. By characterizing the JMJD2B binding motifs and other transcription factor binding datasets, we discover that JMJD2B forms a protein complex with AP-2 family transcription factor TFAP2C and histone demethylase LSD1. The JMJD2B-TFAP2C-LSD1 complex predominantly occupies active gene promoters, whereas the TFAP2C-LSD1 complex is located at putative enhancers, suggesting that these proteins mediate enhancer-promoter interaction for gene regulation. We conclude that JMJD2B is vital to the TSC transcriptional program and safeguards the trophoblast cell fate via distinctive protein interactors and epigenetic targets.


Subject(s)
Epigenesis, Genetic/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Trophoblasts/metabolism , Adult Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Chromatin Immunoprecipitation Sequencing/methods , Epigenomics/methods , Gene Expression , Gene Expression Profiling/methods , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histone Demethylases/physiology , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/physiology , Mice , Mice, 129 Strain , Promoter Regions, Genetic , Stem Cells/metabolism , Transcription Factor AP-2/metabolism , Transcription, Genetic/genetics , Trophoblasts/physiology
5.
Int J Mol Sci ; 22(2)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33478063

ABSTRACT

Cancer is a major cause of death worldwide. Epigenetic changes in response to external (diet, sports activities, etc.) and internal events are increasingly implicated in tumor initiation and progression. In this review, we focused on post-translational changes in histones and, more particularly, the tri methylation of lysine from histone 3 (H3K27me3) mark, a repressive epigenetic mark often under- or overexpressed in a wide range of cancers. Two actors regulate H3K27 methylation: Jumonji Domain-Containing Protein 3 demethylase (JMJD3) and Enhancer of zeste homolog 2 (EZH2) methyltransferase. A number of studies have highlighted the deregulation of these actors, which is why this scientific review will focus on the role of JMJD3 and, consequently, H3K27me3 in cancer development. Data on JMJD3's involvement in cancer are classified by cancer type: nervous system, prostate, blood, colorectal, breast, lung, liver, ovarian, and gastric cancers.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/physiology , Neoplasms/genetics , Animals , DNA Methylation/genetics , Enhancer of Zeste Homolog 2 Protein/physiology , Epigenesis, Genetic/genetics , Female , Histone Demethylases/physiology , Histones/metabolism , Humans , Male , Neoplasms/metabolism , Neoplasms/pathology
6.
J Neurotrauma ; 38(3): 353-364, 2021 02.
Article in English | MEDLINE | ID: mdl-32977735

ABSTRACT

Neural stem cells (NSCs) play vital roles in the homeostasis of neurological function. Ubiquitously transcribed tetratricopeptide repeat, X chromosome (UTX) is an important regulator of stem cell phenotypes. In our current study, we aimed to investigate whether the conditional knockout of UTX on neural stem cells alters macrophage assembly in response to spinal cord injury (SCI). Conditional knockout Utx of NSC (Utx-KO) mice was used to generate SCI models by the modified Allen method. We reported that neurological function and scar hyperplasia significantly improved in Utx-KO mice after SCI, accompanied by significantly reduced assembly of macrophages. With a 45-fold pathway array and Western blot, we found that Utx-KO could significantly inhibit NF-κB signaling activation and promote the synthesis and secretion of macrophage migration inhibitory factor (MIF) in NSCs. Administration of the selective NF-κB p65 activator betulinic acid and the selective MIF inhibitor ISO-1 confirmed that the activation of NF-κB p65 phosphorylation or inhibition of MIF could eliminate the benefits of Utx-KO in SCI, such as inhibition of macrophage aggregation and reduction in scar proliferation. This study confirmed that UTX in NSCs could alter macrophage migration and improve neurological function recovery after SCI in mice.


Subject(s)
Histone Demethylases/physiology , Macrophages/physiology , NF-kappa B/physiology , Neural Stem Cells/metabolism , Signal Transduction/physiology , Spinal Cord Injuries/pathology , Animals , Cell Movement , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spinal Cord Injuries/etiology , Spinal Cord Injuries/metabolism
7.
Blood ; 137(7): 908-922, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33174606

ABSTRACT

Epigenetic regulation is essential for the maintenance of the hematopoietic system, and its deregulation is implicated in hematopoietic disorders. In this study, UTX, a demethylase for lysine 27 on histone H3 (H3K27) and a component of COMPASS-like and SWI/SNF complexes, played an essential role in the hematopoietic system by globally regulating aging-associated genes. Utx-deficient (UtxΔ/Δ) mice exhibited myeloid skewing with dysplasia, extramedullary hematopoiesis, impaired hematopoietic reconstituting ability, and increased susceptibility to leukemia, which are the hallmarks of hematopoietic aging. RNA-sequencing (RNA-seq) analysis revealed that Utx deficiency converted the gene expression profiles of young hematopoietic stem-progenitor cells (HSPCs) to those of aged HSPCs. Utx expression in hematopoietic stem cells declined with age, and UtxΔ/Δ HSPCs exhibited increased expression of an aging-associated marker, accumulation of reactive oxygen species, and impaired repair of DNA double-strand breaks. Pathway and chromatin immunoprecipitation analyses coupled with RNA-seq data indicated that UTX contributed to hematopoietic homeostasis mainly by maintaining the expression of genes downregulated with aging via demethylase-dependent and -independent epigenetic programming. Of note, comparison of pathway changes in UtxΔ/Δ HSPCs, aged muscle stem cells, aged fibroblasts, and aged induced neurons showed substantial overlap, strongly suggesting common aging mechanisms among different tissue stem cells.


Subject(s)
Aging/genetics , Gene Expression Regulation/genetics , Hematopoiesis/genetics , Hematopoietic System/physiology , Histone Code/genetics , Histone Demethylases/physiology , Animals , Cellular Senescence/genetics , DNA Breaks, Double-Stranded , DNA Repair , Female , Genetic Predisposition to Disease , Hematopoiesis, Extramedullary , Histone Demethylases/deficiency , Histone Demethylases/genetics , Immune Reconstitution , Jumonji Domain-Containing Histone Demethylases/metabolism , Leukemia, Experimental/genetics , Leukemia, Experimental/virology , Male , Mice , Mice, Knockout , Moloney murine leukemia virus/physiology , Myeloid Cells/pathology , Radiation Chimera , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Transcription Factors/metabolism , Virus Integration
8.
Plant Physiol Biochem ; 158: 420-433, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33257231

ABSTRACT

The jumonji C (JMJ-C) domain-containing protein is a histone demethylase and is involved in plant stress. However, the function of the JMJ-C gene family in cotton is still not confirmed. Herein, 25, 26, 52, and 53 members belonging to the JMJ-C gene family were identified in Gossypium raimondii, Gossypium arboreum, Gossypium hirsutum, and Gossypium barbadense, respectively. Based on phylogenetic relationships and conserved domains, the JMJ-C genes were categorized into five subfamilies, KDM3, KDM4, KDM5, JMJC, and JMJD6. The chromosomal location, gene structure, motif compositions, and cis-elements have been displayed. The collinear investigation showed that whole-genome duplication event is the mainly power to drive JMJ-C gene family expansion. Transcriptome and qRT-PCR analysis revealed that eight GhJMJs were induced by salt and PEG treatment. Further assays confirmed that GhJMJ34/40 greatly improved salt and osmotic tolerance in Saccharomyces cerevisiae. These results help clarify JMJ-C protein functions in preparation for further study.


Subject(s)
Gossypium/enzymology , Histone Demethylases/physiology , Plant Proteins/physiology , Salt Tolerance , Gene Expression Regulation, Plant , Genome, Plant , Gossypium/genetics , Histone Demethylases/genetics , Multigene Family , Phylogeny , Plant Proteins/genetics
9.
J Biomed Nanotechnol ; 16(6): 899-909, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-33187585

ABSTRACT

A well-studied subject of epigenetics, the histone methylation located at lysine and arginine is overseen via methyltransferases and demethylases. Lysine-specific demethylase 4A (KDM4A) comprises a lysine demethylase and possesses specificity for H3K9me3 and H3K36me3, which is capable of being used in order to activate histone transcription. Our team examined the expression of KDM4A within Sprague Dawley (SD) rats and further investigated the mechanism via which this phenomena regulates osteogenic variation within the present study. The overexpression of KDM4A facilitated the process of osteoblast differentiation in bone mesenchymal stem cells (BMSC), while the knocking down differentiation via osteoblast was restrained via the suppression of the expression of Runx2, Osterix, alkaline phosphatase (ALP), and osteocalcin (OCN). Knocking down KDM4A lowered levels of the promoter expression of Runx2, osterix, and OCN, and raised levels of H3K27me3 expression. The results demonstrated that KDM4A possesses a crucial role within the differentiation of osteoblasts and furthermore regulates the expression of Runx2, Osterix, and OCN via H3K9me3. The present research may provide new insights into the treatment of bone healing.


Subject(s)
Histone Demethylases , Lysine , Osteogenesis , Animals , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/genetics , Histone Demethylases/physiology , Osteoblasts/metabolism , Osteocalcin/genetics , Osteogenesis/genetics , Rats , Rats, Sprague-Dawley , Receptors, Oxytocin , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Int J Mol Sci ; 21(17)2020 Aug 24.
Article in English | MEDLINE | ID: mdl-32847068

ABSTRACT

Kidney cancer is one of the most difficult cancers to treat by targeted and radiation therapy. Therefore, identifying key regulators in this cancer is especially important for finding new drugs. We focused on androgen receptor (AR) regulation by its epigenetic co-regulator lysine-specific histone demethylase 1 (LSD1) in kidney cancer development. LSD1 knock-down in kidney cancer cells decreased expression of AR target genes. Moreover, the binding of AR to target gene promoters was reduced and histone methylation status was changed in LSD1 knock-down kidney cancer cells. LSD1 knock-down also slowed growth and decreased the migration ability of kidney cancer cells. We found that pargyline, known as a LSD1 inhibitor, can reduce AR activity in kidney cancer cells. The treatment of kidney cancer cells with pargyline delayed growth and repressed epithelial-mesenchymal transition (EMT) markers. These effects were additively enhanced by co-treatment with the AR inhibitor enzalutamide. Down-regulation of LSD1 in renal cancer cells (RCC) attenuated in vivo tumor growth in a xenograft mouse model. These results provide evidence that LSD1 can regulate kidney cancer cell growth via epigenetic control of AR transcription factors and that LSD1 inhibitors may be good candidate drugs for treating kidney cancer.


Subject(s)
Carcinoma, Renal Cell/pathology , Histone Demethylases/physiology , Kidney Neoplasms/pathology , Receptors, Androgen/metabolism , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cells, Cultured , Disease Progression , Gene Expression Regulation, Neoplastic , HEK293 Cells , Histone Demethylases/genetics , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Androgen/genetics , Signal Transduction/genetics
11.
Nucleic Acids Res ; 48(16): 8943-8958, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32697292

ABSTRACT

The epithelial-to-mesenchymal transition (EMT) is a complex transcriptional program induced by transforming growth factor ß1 (TGF-ß1). Histone lysine-specific demethylase 1 (LSD1) has been recognized as a key mediator of EMT in cancer cells, but the precise mechanism that underlies the activation and repression of EMT genes still remains elusive. Here, we characterized the early events induced by TGF-ß1 during EMT initiation and establishment. TGF-ß1 triggered, 30-90 min post-treatment, a nuclear oxidative wave throughout the genome, documented by confocal microscopy and mass spectrometry, mediated by LSD1. LSD1 was recruited with phosphorylated SMAD2/3 to the promoters of prototypic genes activated and repressed by TGF-ß1. After 90 min, phospho-SMAD2/3 downregulation reduced the complex and LSD1 was then recruited with the newly synthesized SNAI1 and repressors, NCoR1 and HDAC3, to the promoters of TGF-ß1-repressed genes such as the Wnt soluble inhibitor factor 1 gene (WIF1), a change that induced a late oxidative burst. However, TGF-ß1 early (90 min) repression of transcription also required synchronous signaling by reactive oxygen species and the stress-activated kinase c-Jun N-terminal kinase. These data elucidate the early events elicited by TGF-ß1 and the priming role of DNA oxidation that marks TGF-ß1-induced and -repressed genes involved in the EMT.


Subject(s)
DNA/metabolism , Epithelial-Mesenchymal Transition/genetics , Histone Demethylases/physiology , Smad2 Protein/physiology , Transforming Growth Factor beta1/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans
12.
Nat Plants ; 6(7): 823-837, 2020 07.
Article in English | MEDLINE | ID: mdl-32572214

ABSTRACT

Histone demethylation is crucial for proper chromatin structure and to ensure normal development, and requires the large family of Jumonji C (JmjC)-containing demethylases; however, the molecular mechanisms that regulate the substrate specificity of these JmjC-containing demethylases remain largely unknown. Here, we show that the substrate specificity of the Arabidopsis histone demethylase JMJ16 is broadened from Lys 4 of histone H3 (H3K4) alone in somatic cells to both H3K4 and H3K9 when it binds to the meiocyte-specific histone reader MMD1. Consistent with this, the JMJ16 catalytic domain exhibits both H3K4 and H3K9 demethylation activities. Moreover, the JMJ16 C-terminal FYR domain interacts with the JMJ16 catalytic domain and probably restricts its substrate specificity. By contrast, MMD1 can compete with the N-terminal catalytic domain of JMJ16 for binding to the FYR-C domain, thereby expanding the substrate specificity of JMJ16 by preventing the FYR domain from binding to the catalytic domain. We propose that MMD1 and JMJ16 together in male meiocytes promote gene expression in an H3K9me3-dependent manner and thereby contribute to meiotic chromosome condensation.


Subject(s)
Arabidopsis Proteins/physiology , Arabidopsis/enzymology , Chromosomes, Plant/metabolism , Histone Demethylases/physiology , Meiosis , Arabidopsis/physiology , Arabidopsis Proteins/metabolism , Catalytic Domain , Epigenesis, Genetic , Gene Expression Regulation, Plant , Histone Demethylases/metabolism , Meiosis/physiology , Substrate Specificity
13.
Nucleic Acids Res ; 48(9): 4915-4927, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32232336

ABSTRACT

Post-translational histone modifications and chromatin remodelling play a critical role controlling the integrity of the genome. Here, we identify histone lysine demethylase PHF2 as a novel regulator of the DNA damage response by regulating DNA damage-induced focus formation of 53BP1 and BRCA1, critical factors in the pathway choice for DNA double strand break repair. PHF2 knockdown leads to impaired BRCA1 focus formation and delays the resolution of 53BP1 foci. Moreover, irradiation-induced RPA phosphorylation and focus formation, as well as localization of CtIP, required for DNA end resection, to sites of DNA lesions are affected by depletion of PHF2. These results are indicative of a defective resection of double strand breaks and thereby an impaired homologous recombination upon PHF2 depletion. In accordance with these data, Rad51 focus formation and homology-directed double strand break repair is inhibited in cells depleted for PHF2. Importantly, we demonstrate that PHF2 knockdown decreases CtIP and BRCA1 protein and mRNA levels, an effect that is dependent on the demethylase activity of PHF2. Furthermore, PHF2-depleted cells display genome instability and are mildly sensitive to the inhibition of PARP. Together these results demonstrate that PHF2 promotes DNA repair by homologous recombination by controlling CtIP-dependent resection of double strand breaks.


Subject(s)
DNA Breaks, Double-Stranded , Histone Demethylases/physiology , Homeodomain Proteins/physiology , Recombinational DNA Repair , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Line , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Gene Expression Regulation , Genomic Instability , HeLa Cells , Histone Demethylases/metabolism , Homeodomain Proteins/metabolism , Humans
14.
Clin Cancer Res ; 26(8): 2065-2079, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32047002

ABSTRACT

PURPOSE: Epigenetic deregulation is deeply implicated in the pathogenesis of bladder cancer. KDM6A (Lysine (K)-specific demethylase 6A) is a histone modifier frequently mutated in bladder cancer. However, the molecular mechanisms of how KDM6A deficiency contributes to bladder cancer development remains largely unknown. We hypothesized that clarification of the pathogenic mechanisms underlying KDM6A-mutated bladder cancer can help in designing new anticancer therapies. EXPERIMENTAL DESIGN: We generated mice lacking Kdm6a in the urothelium and crossed them with mice heterozygous for p53, whose mutation/deletion significantly overlaps with the KDM6A mutation in muscle-invasive bladder cancer (MIBC). In addition, BBN (N-butyl-N-(4-hydroxybutyl) nitrosamine), a cigarette smoke-like mutagen, was used as a tumor-promoting agent. Isolated urothelia were subjected to phenotypic, pathologic, molecular, and cellular analyses. The clinical relevance of our findings was further analyzed using genomic and clinical data of patients with MIBC. RESULTS: We found that Kdm6a deficiency activated cytokine and chemokine pathways, promoted M2 macrophage polarization, increased cancer stem cells and caused bladder cancer in cooperation with p53 haploinsufficiency. We also found that BBN treatment significantly enhanced the expression of proinflammatory molecules and accelerated disease development. Human bladder cancer samples with decreased KDM6A expression also showed activated proinflammatory pathways. Notably, dual inhibition of IL6 and chemokine (C-C motif) ligand 2, upregulated in response to Kdm6a deficiency, efficiently suppressed Kdm6a-deficient bladder cancer cell growth. CONCLUSIONS: Our findings provide insights into multistep carcinogenic processes of bladder cancer and suggest molecular targeted therapeutic approaches for patients with bladder cancer with KDM6A dysfunction.


Subject(s)
Carcinogenesis/pathology , Histone Demethylases/physiology , Inflammation/pathology , Macrophages/immunology , Tumor Suppressor Protein p53/physiology , Urinary Bladder Neoplasms/pathology , Urothelium/pathology , Animals , Carcinogenesis/genetics , Carcinogenesis/immunology , Databases, Genetic/statistics & numerical data , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Mice , Mice, Inbred C3H , Mice, Knockout , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/metabolism , Urothelium/immunology
15.
Metabolism ; 102: 154011, 2020 01.
Article in English | MEDLINE | ID: mdl-31734274

ABSTRACT

OBJECTIVE: The gut microbiota regulates thermogenesis to benefit metabolic homeostasis at least partially via its metabolite butyrate, and the underlying mechanisms of this regulation are still unclear. In this study, we aim to investigate the role of lysine specific demethylase (LSD1), a histone demethylase and important regulator of thermogenesis, in mediating gut microbial metabolite butyrate regulation of thermogenesis. METHODS: The antibiotic cocktail (ABX) was administrated to deplete gut microbiota. Adipose-specific LSD1 knockout mice (LSD1 aKO) were generated by crossing LSD1-lox/lox with adiponectin-cre mice and sodium butyrate and dietary fiber inulin was administrated through oral-gavage. Primary stromal vascular cells were isolated from adipose tissues and differentiated to adipocytes for studying butyrate effects on adipocyte thermogenesis. RESULTS: The antibiotic cocktail (ABX)-mediated depletion of the gut microbiota in mice downregulated the expression of LSD1 in both brown adipose tissue (BAT) and subcutaneous white adipose tissue (scWAT) in addition to uncoupling protein 1 (UCP1) and body temperature. Gavage of the microbial metabolite butyrate in ABX-treated mice reversed the thermogenic functional impairment and LSD1 expression. The adipose-specific ablation of LSD1 in mice attenuated the butyrate-mediated induction of thermogenesis and energy expenditure. Notably, our results showed that butyrate directly increased the expression of LSD1 and UCP1 as well as butyrate transporter monocarboxylate transporter 1 (MCT1) and catabolic enzyme acyl-CoA medium-chain synthetase 3 (ACSM3) in ex vivo cultured adipocytes. The inhibition of MCT1 blocked the effects of butyrate in adipocytes. Furthermore, the butyrate-mediated prevention of diet-induced obesity (DIO) through increased thermogenesis was attenuated in LSD1 aKO mice. Moreover, after gavaging HFD-fed mice with the dietary fiber inulin, a substrate of microbial fermentation that rapidly produces butyrate, thermogenesis in both BAT and scWAT was increased, and DIO was decreased; however, these beneficial metabolic effects were blocked in LSD1 aKO mice. CONCLUSIONS: Together, our results indicate that the microbial metabolite butyrate regulates thermogenesis in BAT and scWAT through the activation of LSD1.


Subject(s)
Adipose Tissue, Brown/drug effects , Adipose Tissue, White/drug effects , Butyrates/pharmacology , Gastrointestinal Microbiome/physiology , Histone Demethylases/physiology , Thermogenesis/drug effects , Thermogenesis/genetics , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Animals , Butyrates/metabolism , Cells, Cultured , Energy Metabolism/drug effects , Energy Metabolism/genetics , Gene Expression Regulation, Enzymologic/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Subcutaneous Fat/metabolism
16.
Development ; 146(24)2019 12 20.
Article in English | MEDLINE | ID: mdl-31862793

ABSTRACT

In Drosophila, the larval prothoracic gland integrates nutritional status with developmental signals to regulate growth and maturation through the secretion of the steroid hormone ecdysone. While the nutritional signals and cellular pathways that regulate prothoracic gland function are relatively well studied, the transcriptional regulators that orchestrate the activity of this tissue remain less characterized. Here, we show that lysine demethylase 5 (KDM5) is essential for prothoracic gland function. Indeed, restoring kdm5 expression only in the prothoracic gland in an otherwise kdm5 null mutant animal is sufficient to rescue both the larval developmental delay and the pupal lethality caused by loss of KDM5. Our studies show that KDM5 functions by promoting the endoreplication of prothoracic gland cells, a process that increases ploidy and is rate limiting for the expression of ecdysone biosynthetic genes. Molecularly, we show that KDM5 activates the expression of the receptor tyrosine kinase torso, which then promotes polyploidization and growth through activation of the MAPK signaling pathway. Taken together, our studies provide key insights into the biological processes regulated by KDM5 and expand our understanding of the transcriptional regulators that coordinate animal development.


Subject(s)
Biological Clocks/genetics , Drosophila Proteins/physiology , Drosophila melanogaster , Embryonic Development/genetics , Endocrine Glands/embryology , Histone Demethylases/physiology , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Ecdysone/metabolism , Embryo, Nonmammalian , Endocrine Glands/metabolism , Endoreduplication/genetics , Female , Gene Expression Regulation, Developmental , Larva , MAP Kinase Signaling System/physiology , Male , Organogenesis/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Time Factors
17.
Cells ; 8(10)2019 10 07.
Article in English | MEDLINE | ID: mdl-31591366

ABSTRACT

: The methylation of histone lysine residues modifies chromatin conformation and regulates the expression of genes implicated in cell metabolism. Lysine-specific demethylase 1 (LSD1) is a flavin-dependent monoamine oxidase that can demethylate mono- and dimethylated histone lysines 4 and 9 (H3K4 and H3K9). The removal of methyl groups from the lysine residues of histone and non-histone proteins was found to be an important regulatory factor of cell proliferation. However, its role has not been fully elucidated. In this study, we assessed LSD1-mediated cell cycle progression using a human endothelial cell model. The short hairpin RNA knockdown of LSD1 inhibits the G2/M phase of cell cycle progression by checkpoint kinase 1 (Chk1) phosphorylation (S137). We observed elevated DNA damage, which was consistent with the increased detection of double-strand breaks as well as purines and pyrimidines oxidation, which accompanied the activation of ATR/ATRIP signaling by H2AXS139 phosphorylation. The irreversible pharmacological inhibition of LSD1 by 2-phenylcyclopropylamine (2-PCPA) inactivated its enzymatic activity, causing significant changes in heterochromatin and euchromatin conformation assessed by chromatin assembly factor 1 subunit A (CAF1A) and heterochromatin protein 1 isoform α and γ (HP1α/γ) immunofluorescence analysis. We conclude that the knockdown of LSD1 in endothelial cells leads to increased HP1-positive chromatin, the stimulation of DNA repair processes, and the dysregulation of proliferation machinery.


Subject(s)
Checkpoint Kinase 1/metabolism , Chromatin/metabolism , Endothelial Cells , Histone Demethylases/physiology , Cell Line , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , DNA Repair , Demethylation , Endothelial Cells/cytology , Endothelial Cells/metabolism , G2 Phase Cell Cycle Checkpoints/physiology , Gene Silencing , Histone Demethylases/genetics , Humans , M Phase Cell Cycle Checkpoints/physiology , Phosphorylation , Protein Processing, Post-Translational
18.
Int J Biol Sci ; 15(11): 2363-2372, 2019.
Article in English | MEDLINE | ID: mdl-31595154

ABSTRACT

Following fertilization, the zygotic genome is activated through a process termed zygotic genome activation (ZGA), which enables zygotic gene products to replace the maternal products and initiates early embryonic development. During the ZGA period, the embryonic epigenome experiences extensive recodifications. The H3K27me3 demethylase UTX is essential for post-implantation embryonic development. However, it remains unclear whether UTX participates in preimplantation development, especially during the ZGA process. In the present study, we showed that either knockdown or overexpression of UTX led to embryonic development retardation, whereas simultaneous depletion of UTX and overexpression of ZSCAN4D rescued the embryonic development, indicating that UTX positively regulated Zscan4d expression. Using a transgenic mice model, we also found that UTX was required for preimplantation embryonic development. In conclusion, these results indicate that UTX functions as a novel regulator and plays critical roles during ZGA in addition to early embryonic development.


Subject(s)
Embryonic Development/genetics , Gene Expression Regulation, Developmental , Histone Demethylases/physiology , Transcription Factors/genetics , Animals , Genome , Histone Demethylases/genetics , Histone Demethylases/metabolism , Mice, Transgenic , Transcription Factors/metabolism , Zygote
19.
Mol Ther ; 27(12): 2134-2146, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31495776

ABSTRACT

The regeneration of the blood vessel system post spinal cord injury (SCI) is essential for the repair of neurological function. As a significant means to regulate gene expression, epigenetic regulation of angiogenesis in SCI is still largely unknown. Here, we found that Ubiquitously Transcribed tetratricopeptide repeat on chromosome X (UTX), the histone H3K27 demethylase, increased significantly in endothelial cells post SCI. Knockdown of UTX can promote the migration and tube formation of endothelial cells. The specific knockout of UTX in endothelial cells enhanced angiogenesis post SCI accompanied with improved neurological function. In addition, we found regulation of UTX expression can change the level of microRNA 24 (miR-24) in vitro. The physical binding of UTX to the promotor of miR-24 was indicated by chromatin immunoprecipitation (ChIP) assay. Meanwhile, methylation sequencing of endothelial cells demonstrated that UTX could significantly decrease the level of methylation in the miR-24 promotor. Furthermore, miR-24 significantly abolished the promoting effect of UTX deletion on angiogenesis in vitro and in vivo. Finally, we predicted the potential target mRNAs of miR-24 related to angiogenesis. We indicate that UTX deletion can epigenetically promote the vascular regeneration and functional recovery post SCI by forming a regulatory network with miR-24.


Subject(s)
Endothelial Cells/cytology , Epigenesis, Genetic , Gene Deletion , Histone Demethylases/physiology , Regeneration , Spinal Cord Injuries/therapy , Animals , Cell Movement , Cell Proliferation , DNA Methylation , Endothelial Cells/physiology , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Promoter Regions, Genetic , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology
20.
Cell Signal ; 64: 109415, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31513837

ABSTRACT

It has been reported that KDM3A participates in several cardiovascular diseases through epigenetic mechanisms. However, its biological role post myocardial infarction (MI) has not been explored. Excessive and prolonged inflammation period can aggravate post-MI injuries and accelerates left ventricular (LV) remodeling. Previous studies have shown that macrophages play a momentous role in post-MI injuries by regulating the balance between the inflammatory phase. In this study, we aimed to demonstrate whether KDM3A could regulate the polarization of macrophages to affect the inflammatory response after myocardial infarction and whether targeting KDM3A could influence the prognosis of myocardial infarction and adverse LV remodeling. To explore the biological function of KDM3A and the underlying mechanisms, the loss of function experiments were designed in vitro and vivo. we analyzed the function of macrophages by a phagocytosis and migration assay and explored the polarization of macrophages. The expression of macrophage inflammation-related genes in the acute inflammatory phase and surface markers was detected by western blot and immunofluorescence assays. Echocardiography, Masson's trichrome staining and hematoxylin and eosin (H&E) staining were used to detect cardiac ventricular function. Our data showed that KDM3A is essential for the biological function of rat bone marrow macrophages (BMDMs), and KDM3A deficiency decreases the capacity for phagocytosis and migration, promoting M1 but restraining M2 macrophage phenotype polarization in vitro. Furthermore, we constructed MI models of male rats to verify that KDM3A deficiency could regulate macrophage polarization to aggravate the inflammatory response and accelerate LV remodeling in vivo. Among them, we confirmed that IRF4 is a downstream effector of the KDM3A-dependent pathway which could epigenetically influence the transcription of IRF4 by enhancing histone H3 lysine 9 di-methylation(H3K9me2) accumulation on the IRF4 gene proximal promoter region to modulate macrophage polarization. These results demonstrated that KDM3A plays an essential role in the cardiac repair process of post-MI and LV remodeling by modulating the macrophage phenotype, thereby suggesting a promising therapy to treat post MI injuries.


Subject(s)
Histone Demethylases/physiology , Inflammation/metabolism , Macrophage Activation , Myocardial Infarction/metabolism , Myocardium/metabolism , Ventricular Remodeling , Animals , Cells, Cultured , Disease Models, Animal , Histone Demethylases/antagonists & inhibitors , Interferon Regulatory Factors/metabolism , Macrophages , Male , Myocardial Infarction/pathology , Myocardium/pathology , Phagocytosis , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL