Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Elife ; 122023 01 25.
Article in English | MEDLINE | ID: mdl-36695573

ABSTRACT

Skeletal muscle exhibits remarkable plasticity in response to environmental cues, with stress-dependent effects on the fast-twitch and slow-twitch fibers. Although stress-induced gene expression underlies environmental adaptation, it is unclear how transcriptional and epigenetic factors regulate fiber type-specific responses in the muscle. Here, we show that flavin-dependent lysine-specific demethylase-1 (LSD1) differentially controls responses to glucocorticoid and exercise in postnatal skeletal muscle. Using skeletal muscle-specific LSD1-knockout mice and in vitro approaches, we found that LSD1 loss exacerbated glucocorticoid-induced atrophy in the fast fiber-dominant muscles, with reduced nuclear retention of Foxk1, an anti-autophagic transcription factor. Furthermore, LSD1 depletion enhanced endurance exercise-induced hypertrophy in the slow fiber-dominant muscles, by induced expression of ERRγ, a transcription factor that promotes oxidative metabolism genes. Thus, LSD1 serves as an 'epigenetic barrier' that optimizes fiber type-specific responses and muscle mass under the stress conditions. Our results uncover that LSD1 modulators provide emerging therapeutic and preventive strategies against stress-induced myopathies such as sarcopenia, cachexia, and disuse atrophy.


Subject(s)
Glucocorticoids , Muscular Diseases , Mice , Animals , Glucocorticoids/metabolism , Muscle, Skeletal/metabolism , Muscle Fibers, Skeletal/metabolism , Transcription Factors/metabolism , Histone Demethylases/genetics , Histone Demethylases/metabolism
2.
Blood Adv ; 5(9): 2305-2318, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33929501

ABSTRACT

Acute myeloid leukemia (AML) is a heterogenous malignancy characterized by distinct lineage subtypes and various genetic/epigenetic alterations. As with other neoplasms, AML cells have well-known aerobic glycolysis, but metabolic variations depending on cellular lineages also exist. Lysine-specific demethylase-1 (LSD1) has been reported to be crucial for human leukemogenesis, which is currently one of the emerging therapeutic targets. However, metabolic roles of LSD1 and lineage-dependent factors remain to be elucidated in AML cells. Here, we show that LSD1 directs a hematopoietic lineage-specific metabolic program in AML subtypes. Erythroid leukemia (EL) cells particularly showed activated glycolysis and high expression of LSD1 in both AML cell lines and clinical samples. Transcriptome, chromatin immunoprecipitation-sequencing, and metabolomic analyses revealed that LSD1 was essential not only for glycolysis but also for heme synthesis, the most characteristic metabolic pathway of erythroid origin. Notably, LSD1 stabilized the erythroid transcription factor GATA1, which directly enhanced the expression of glycolysis and heme synthesis genes. In contrast, LSD1 epigenetically downregulated the granulo-monocytic transcription factor C/EBPα. Thus, the use of LSD1 knockdown or chemical inhibitor dominated C/EBPα instead of GATA1 in EL cells, resulting in metabolic shifts and growth arrest. Furthermore, GATA1 suppressed the gene encoding C/EBPα that then acted as a repressor of GATA1 target genes. Collectively, we conclude that LSD1 shapes metabolic phenotypes in EL cells by balancing these lineage-specific transcription factors and that LSD1 inhibitors pharmacologically cause lineage-dependent metabolic remodeling.


Subject(s)
Leukemia, Erythroblastic, Acute , CCAAT-Enhancer-Binding Protein-alpha , GATA1 Transcription Factor/genetics , Histone Demethylases/genetics , Humans , Leukemia, Erythroblastic, Acute/genetics , Proto-Oncogene Proteins , Transcription Factors
3.
Trends Endocrinol Metab ; 30(7): 409-412, 2019 07.
Article in English | MEDLINE | ID: mdl-31151734

ABSTRACT

Various nutritional signals are transduced by two epigenetic pathways: NAD-dependent sirtuin Sirt1 (NAD+-Sirt1) deacetylase and flavin adenine dinucleotide-dependent lysine-specific demethylase 1 (FAD-LSD1). These pathways are controlled by dietary vitamins and nutrient-responsive hormones such as glucocorticoids and insulin, resulting in endocrine-metabolism-epigenome cooperation in adipocyte and skeletal muscle development.


Subject(s)
Flavin-Adenine Dinucleotide/metabolism , Histone Demethylases/metabolism , NAD/metabolism , Sirtuin 1/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Flavin-Adenine Dinucleotide/genetics , Glucocorticoids/metabolism , Histone Demethylases/genetics , Humans , NAD/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Sirtuin 1/genetics
4.
FASEB J ; 33(4): 5300-5311, 2019 04.
Article in English | MEDLINE | ID: mdl-30681884

ABSTRACT

Transcriptional and epigenetic regulation is fundamentally involved in initiating and maintaining progression of cellular differentiation. The 2 types of thermogenic adipocytes, brown and beige, are thought to be of different origins but share functionally similar phenotypes. Here, we report that lysine-specific demethylase 2 (LSD2) regulates the expression of genes associated with lineage identity during the differentiation of brown and beige adipogenic progenitors in mice. In HB2 mouse brown preadipocytes, short hairpin RNA-mediated knockdown (KD) of LSD2 impaired formation of lipid droplet-containing adipocytes and down-regulated brown adipogenesis-associated genes. Transcriptomic analysis revealed that myogenesis-associated genes were up-regulated in LSD2-KD cells under adipogenic induction. In addition, loss of LSD2 during later phases of differentiation had no obvious influence on adipogenic traits, suggesting that LSD2 functions during earlier phases of brown adipocyte differentiation. Using adipogenic cells from the brown adipose tissues of LSD2-knockout (KO) mice, we found reduced expression of brown adipogenesis genes, whereas myogenesis genes were not affected. In contrast, when LSD2-KO cells from inguinal white adipose tissues were subjected to beige induction, these cells showed a dramatic rise in myogenic gene expression. Collectively, these results suggest that LSD2 regulates distinct sets of genes during brown and beige adipocyte formation.-Takase, R., Hino, S., Nagaoka, K., Anan, K., Kohrogi, K., Araki, H., Hino, Y., Sakamoto, A., Nicholson, T. B., Chen, T., Nakao, M. Lysine-specific demethylase-2 is distinctively involved in brown and beige adipogenic differentiation.


Subject(s)
Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Histone Demethylases/metabolism , RNA, Small Interfering/metabolism , Adipogenesis/genetics , Adipogenesis/physiology , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Chromatin Immunoprecipitation , Female , Histone Demethylases/genetics , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/metabolism , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction
5.
Nucleic Acids Res ; 46(11): 5441-5454, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29618057

ABSTRACT

The metabolic properties of cells are formed under the influence of environmental factors such as nutrients and hormones. Although such a metabolic program is likely initiated through epigenetic mechanisms, the direct links between metabolic cues and activities of chromatin modifiers remain largely unknown. In this study, we show that lysine-specific demethylase-1 (LSD1) controls the metabolic program in myogenic differentiation, under the action of catabolic hormone, glucocorticoids. By using transcriptomic and epigenomic approaches, we revealed that LSD1 bound to oxidative metabolism and slow-twitch myosin genes, and repressed their expression. Consistent with this, loss of LSD1 activity during differentiation enhanced the oxidative capacity of myotubes. By testing the effects of various hormones, we found that LSD1 levels were decreased by treatment with the glucocorticoid dexamethasone (Dex) in cultured myoblasts and in skeletal muscle from mice. Mechanistically, glucocorticoid signaling induced expression of a ubiquitin E3 ligase, JADE-2, which was responsible for proteasomal degradation of LSD1. Consequently, in differentiating myoblasts, chemical inhibition of LSD1, in combination with Dex treatment, synergistically de-repressed oxidative metabolism genes, concomitant with increased histone H3 lysine 4 methylation at these loci. These findings demonstrated that LSD1 serves as an epigenetic regulator linking glucocorticoid action to metabolic programming during myogenic differentiation.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Histone Demethylases/metabolism , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/cytology , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Histone Demethylases/antagonists & inhibitors , Histones/metabolism , Male , Methylation , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Oxidation-Reduction , Ubiquitin-Protein Ligases/metabolism
6.
Cancer Res ; 75(7): 1445-56, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25649769

ABSTRACT

The hallmark of most cancer cells is the metabolic shift from mitochondrial to glycolytic metabolism for adapting to the surrounding environment. Although epigenetic modification is intimately linked to cancer, the molecular mechanism, by which epigenetic factors regulate cancer metabolism, is poorly understood. Here, we show that lysine-specific demethylase-1 (LSD1, KDM1A) has an essential role in maintaining the metabolic shift in human hepatocellular carcinoma cells. Inhibition of LSD1 reduced glucose uptake and glycolytic activity, with a concurrent activation of mitochondrial respiration. These metabolic changes coexisted with the inactivation of the hypoxia-inducible factor HIF1α, resulting in a decreased expression of GLUT1 and glycolytic enzymes. In contrast, during LSD1 inhibition, a set of mitochondrial metabolism genes was activated with the concomitant increase of methylated histone H3 at lysine 4 in the promoter regions. Consistently, both LSD1 and GLUT1 were significantly overexpressed in carcinoma tissues. These findings demonstrate the epigenetic plasticity of cancer cell metabolism, which involves an LSD1-mediated mechanism.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Histone Demethylases/physiology , Liver Neoplasms/enzymology , Mitochondria, Liver/enzymology , Animals , Carcinoma, Hepatocellular/pathology , Gene Expression , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glycolysis , Hep G2 Cells , Histones/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Liver Neoplasms/pathology , Methylation , Mice, SCID , Neoplasm Transplantation , Protein Processing, Post-Translational , Tumor Burden
7.
Mol Cell Biol ; 35(7): 1068-80, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25624347

ABSTRACT

Cells link environmental fluctuations, such as nutrition, to metabolic remodeling. Epigenetic factors are thought to be involved in such cellular processes, but the molecular basis remains unclear. Here we report that the lysine-specific demethylase 2 (LSD2) suppresses the flux and metabolism of lipids to maintain the energy balance in hepatic cells. Using transcriptome and chromatin immunoprecipitation-sequencing analyses, we revealed that LSD2 represses the genes involved in lipid influx and metabolism through demethylation of histone H3K4. Selective recruitment of LSD2 at lipid metabolism gene loci was mediated in part by a stress-responsive transcription factor, c-Jun. Intriguingly, LSD2 depletion increased the intracellular levels of many lipid metabolites, which was accompanied by an increased susceptibility to toxic cell damage in response to fatty acid exposure. Our data demonstrate that LSD2 maintains metabolic plasticity under fluctuating environment in hepatocytes by mediating the cross talk between the epigenome and metabolism.


Subject(s)
Down-Regulation , Hepatocytes/metabolism , Histone Demethylases/metabolism , Lipid Metabolism , Lipids/genetics , Animals , Hep G2 Cells , Histones/metabolism , Humans , Liver/metabolism , Lysine/metabolism , Male , Methylation , Mice, Inbred C57BL , Proto-Oncogene Proteins c-jun/metabolism
8.
Nat Commun ; 3: 758, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22453831

ABSTRACT

Environmental factors such as nutritional state may act on the epigenome that consequently contributes to the metabolic adaptation of cells and the organisms. The lysine-specific demethylase-1 (LSD1) is a unique nuclear protein that utilizes flavin adenosine dinucleotide (FAD) as a cofactor. Here we show that LSD1 epigenetically regulates energy-expenditure genes in adipocytes depending on the cellular FAD availability. We find that the loss of LSD1 function, either by short interfering RNA or by selective inhibitors in adipocytes, induces a number of regulators of energy expenditure and mitochondrial metabolism such as PPARγ coactivator-1α resulting in the activation of mitochondrial respiration. In the adipose tissues from mice on a high-fat diet, expression of LSD1-target genes is reduced, compared with that in tissues from mice on a normal diet, which can be reverted by suppressing LSD1 function. Our data suggest a novel mechanism where LSD1 regulates cellular energy balance through coupling with cellular FAD biosynthesis.


Subject(s)
Energy Metabolism , Flavin-Adenine Dinucleotide/metabolism , Mitochondria/metabolism , Oxidoreductases, N-Demethylating/metabolism , 3T3 Cells , Adipocytes/metabolism , Animals , Cell Line , Diet, High-Fat , Eating , Gene Expression Profiling , HEK293 Cells , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histone Demethylases , Humans , Lysine/metabolism , Mice , Mice, Inbred C57BL , Oxidoreductases, N-Demethylating/genetics , PPAR gamma/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , RNA Interference , RNA, Small Interfering , RNA-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...