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.
JCI Insight ; 8(21)2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37937648

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a leading cause for chronic liver diseases. Current therapeutic options are limited due to an incomplete mechanistic understanding of how steatosis transitions to NASH. Here we show that the TRIM21 E3 ubiquitin ligase is induced by the synergistic actions of proinflammatory TNF-α and fatty acids in livers of humans and mice with NASH. TRIM21 ubiquitinates and degrades ChREBP, SREBP1, ACC1, and FASN, key regulators of de novo lipogenesis, and A1CF, an alternative splicing regulator of the high-activity ketohexokinase-C (KHK-C) isoform and rate-limiting enzyme of fructose metabolism. TRIM21-mediated degradation of these lipogenic activators improved steatosis and hyperglycemia as well as fructose and glucose tolerance. Our study identifies TRIM21 as a negative regulator of liver steatosis in NASH and provides mechanistic insights into an immunometabolic crosstalk that limits fatty acid synthesis and fructose metabolism during metabolic stress. Thus, enhancing this natural counteracting force of steatosis through inhibition of key lipogenic activators via TRIM21-mediated ubiquitination may provide a therapeutic opportunity to treat NASH.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Humans , Mice , Fructose/metabolism , Inflammation , Lipogenesis , Ubiquitination
2.
Cell Rep ; 29(2): 283-300.e8, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31597092

ABSTRACT

The regulation of hepatic gene expression has been extensively studied at the transcriptional level; however, the control of metabolism through posttranscriptional gene regulation by RNA-binding proteins in physiological and disease states is less understood. Here, we report a major role for the hormone-sensitive RNA-binding protein (RBP) APOBEC1 complementation factor (A1CF) in the generation of hepatocyte-specific and alternatively spliced transcripts. Among these transcripts are isoforms for the dominant and high-affinity fructose-metabolizing ketohexokinase C and glycerol kinase, two key metabolic enzymes that are linked to hepatic gluconeogenesis and found to be markedly reduced upon hepatic ablation of A1cf. Consequently, mice lacking A1CF exhibit improved glucose tolerance and are protected from fructose-induced hyperglycemia, hepatic steatosis, and development of obesity. Our results identify a previously unreported function of A1CF as a regulator of alternative splicing of a subset of genes influencing hepatic glucose production through fructose and glycerol metabolism.


Subject(s)
Alternative Splicing/genetics , Fructose/metabolism , Glycerol/metabolism , Liver/metabolism , RNA-Binding Proteins/metabolism , RNA/genetics , 3' Untranslated Regions/genetics , Animals , Base Sequence , Cell Line, Tumor , Fatty Liver/genetics , Genome , Gluconeogenesis , Homeostasis , Humans , Hyperglycemia/genetics , Insulin/metabolism , Introns/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Protein Transport , RNA Splice Sites/genetics , Subcellular Fractions/metabolism , Transcriptome/genetics
3.
Cell Rep ; 20(4): 909-922, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28746875

ABSTRACT

H4K20 monomethylation maintains genome integrity by regulating proper mitotic condensation, DNA damage response, and replication licensing. Here, we show that, in non-dividing hepatic cells, H4K20Me1 is specifically enriched in active gene bodies and dynamically regulated by the antagonistic action of Kmt5a methylase and Kdm7b demethylase. In liver-specific Kmt5a-deficient mice, reduced levels of H4K20Me1 correlated with reduced RNA Pol II release from promoter-proximal regions. Genes regulating glucose and fatty acid metabolism were most sensitive to impairment of RNA Pol II release. Downregulation of glycolytic genes resulted in an energy starvation condition partially compensated by AMP-activated protein kinase (AMPK) activation and increased mitochondrial activity. This metabolic reprogramming generated a highly sensitized state that, upon different metabolic stress conditions, quickly aggravated into a senescent phenotype due to ROS overproduction-mediated oxidative DNA damage. The results illustrate how defects in the general process of RNA Pol II transition into a productive elongation phase can trigger specific metabolic changes and genome instability.


Subject(s)
Histones/metabolism , Liver/metabolism , Protein Methyltransferases/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic/genetics , Animals , Gene Expression Regulation , Histones/genetics , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protein Methyltransferases/genetics , RNA Polymerase II/genetics
4.
Mol Cell Oncol ; 3(1): e1021946, 2016.
Article in English | MEDLINE | ID: mdl-27308536

ABSTRACT

Cancer stem cells (CSCs) are defined as cells within tumors that can self-renew and differentiate into heterogeneous lineages of cancerous cells. The origin of CSCs is not well understood. Recent evidence suggests that CSCs in hepatocellular carcinoma could be generated via oncogenic transformation and partial differentiation of adult hepatic ductal progenitor cells.

5.
Cell Rep ; 15(12): 2733-44, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27292644

ABSTRACT

TGF-ß signaling regulates a variety of cellular processes, including proliferation, apoptosis, differentiation, immune responses, and fibrogenesis. Here, we describe a lysine methylation-mediated mechanism that controls the pro-fibrogenic activity of TGF-ß. We find that the methyltransferase Set9 potentiates TGF-ß signaling by targeting Smad7, an inhibitory downstream effector. Smad7 methylation promotes interaction with the E3 ligase Arkadia and, thus, ubiquitination-dependent degradation. Depletion or pharmacological inhibition of Set9 results in elevated Smad7 protein levels and inhibits TGF-ß-dependent expression of genes encoding extracellular matrix components. The inhibitory effect of Set9 on TGF-ß-mediated extracellular matrix production is further demonstrated in mouse models of pulmonary fibrosis. Lung fibrosis induced by bleomycin or Ad-TGF-ß treatment was highly compromised in Set9-deficient mice. These results uncover a complex regulatory interplay among multiple Smad7 modifications and highlight the possibility that protein methyltransferases may represent promising therapeutic targets for treating lung fibrosis.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Protein Methyltransferases/metabolism , Pulmonary Fibrosis/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Acetylation , Animals , Bleomycin , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Lysine/metabolism , Male , Methylation , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Nuclear Proteins/metabolism , Protein Stability , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Signal Transduction/genetics , Smad7 Protein/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/genetics
6.
EMBO J ; 34(4): 430-47, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25515659

ABSTRACT

PR-SET7-mediated histone 4 lysine 20 methylation has been implicated in mitotic condensation, DNA damage response and replication licensing. Here, we show that PR-SET7 function in the liver is pivotal for maintaining genome integrity. Hepatocyte-specific deletion of PR-SET7 in mouse embryos resulted in G2 phase arrest followed by massive cell death and defect in liver organogenesis. Inactivation at postnatal stages caused cell duplication-dependent hepatocyte necrosis, accompanied by inflammation, fibrosis and compensatory growth induction of neighboring hepatocytes and resident ductal progenitor cells. Prolonged necrotic regenerative cycles coupled with oncogenic STAT3 activation led to the spontaneous development of hepatic tumors composed of cells with cancer stem cell characteristics. These include a capacity to self-renew in culture or in xenografts and the ability to differentiate to phenotypically distinct hepatic cells. Hepatocellular carcinoma in PR-SET7-deficient mice displays a cancer stem cell gene signature specified by the co-expression of ductal progenitor markers and oncofetal genes.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Neoplastic Stem Cells/enzymology , Animals , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/enzymology , Liver Neoplasms/metabolism , Male , Methylation , Mice , Tumor Cells, Cultured
7.
Clin Cancer Res ; 19(11): 2810-6, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23549874

ABSTRACT

Inflammatory signals from the surrounding microenvironment play important roles in tumor promotion. Key inflammatory mediators and pathways that induce and sustain tumorigenesis have recently been identified in many different cancers. Hepatocellular carcinoma is a paradigm for inflammation-induced cancer, as it most frequently develops in the setting of chronic hepatitis, consecutive cellular damage, and compensatory regeneration. Recent studies revealed that liver damage-mediated inflammation and carcinogenesis are triggered by a complex cross-talk between NF-κB, c-jun-NH2-kinase, and STAT3 signaling pathways. Molecular dissection of the mechanisms involved in the interplay between these pathways identified promising new targets for therapeutic intervention. Targeting different components of the signaling cascades may provide efficient means for blocking the apparently irreversible sequence of events initiated by chronic liver inflammation and culminating in liver cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Signal Transduction , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Humans , Inflammation/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Translational Research, Biomedical
8.
Cancer Cell ; 21(6): 738-50, 2012 Jun 12.
Article in English | MEDLINE | ID: mdl-22698400

ABSTRACT

The tumor suppressor cylindromatosis (CYLD) inhibits the NFκB and mitogen-activated protein kinase (MAPK) activation pathways by deubiquitinating upstream regulatory factors. Here we show that liver-specific disruption of CYLD triggers hepatocyte cell death in the periportal area via spontaneous and chronic activation of TGF-ß activated kinase 1 (TAK1) and c-Jun N-terminal kinase (JNK). This is followed by hepatic stellate cell and Kupffer cell activation, which promotes progressive fibrosis, inflammation, tumor necrosis factor (TNF) production, and expansion of hepatocyte apoptosis toward the central veins. At later stages, compensatory proliferation results in the development of cancer foci featuring re-expression of oncofetal hepatic and stem cell-specific genes. The results demonstrate that, in the liver, CYLD acts as an important regulator of hepatocyte homeostasis, protecting cells from spontaneous apoptosis by preventing uncontrolled TAK1 and JNK activation.


Subject(s)
Apoptosis/genetics , Cysteine Endopeptidases/genetics , Hepatocytes/metabolism , Liver/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Comparative Genomic Hybridization , Cysteine Endopeptidases/metabolism , Deubiquitinating Enzyme CYLD , Enzyme Activation/drug effects , Fibrosis/genetics , Fibrosis/metabolism , Gene Expression Profiling , Hepatocytes/pathology , Immunohistochemistry , Inflammation/genetics , Inflammation/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...