Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 710: 149886, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38581953

ABSTRACT

Mdivi-1, Mitochondrial DIVIsion inhibitor 1, has been widely employed in research under the assumption that it exclusively influences mitochondrial fusion, but effects other than mitochondrial dynamics have been underinvestigated. This paper provides transcriptome and DNA methylome-wide analysis for Mdivi-1 treated SH-SY5Y human neuroblastoma cells using RNA sequencing (RNA-seq) and methyl capture sequencing (MC-seq) methods. Gene ontology analysis of RNA sequences revealed that p53 transcriptional gene network and DNA replication initiation-related genes were significantly up and down-regulated, respectively, showing the correlation with the arrest cell cycle in the G1 phase. MC-seq, a powerful sequencing method for capturing DNA methylation status in CpG sites, revealed that although Mdivi-1 does not induce dramatic DNA methylation change, the subtle alterations were concentrated within the CpG island. Integrative analysis of both sequencing data disclosed that the p53 transcriptional network was activated while the Parkinson's disease pathway was halted. Next, we investigated several changes in mitochondria in response to Mdivi-1. Copy number and transcription of mitochondrial DNA were suppressed. ROS levels increased, and elevated ROS triggered mitochondrial retrograde signaling rather than inducing direct DNA damage. In this study, we could better understand the molecular network of Mdivi-1 by analyzing DNA methylation and mRNA transcription in the nucleus and further investigating various changes in mitochondria, providing inspiration for studying nuclear-mitochondrial communications.


Subject(s)
Dynamins , Neuroblastoma , Humans , Dynamins/metabolism , Mitochondrial Dynamics , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Quinazolinones/pharmacology
2.
Bone ; 177: 116918, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37739296

ABSTRACT

Recently improved techniques could provide snapshots of chromatin structure generated based on chromatin accessibility. Since chromatin accessibility determines transcriptional potential, it has been attempted in a variety of cell systems. However, there has been no genome-wide analysis of chromatin accessibility for the entire murine osteoclast (OC) differentiation process. We performed an Assay for Transposase-Accessible Chromatin (ATAC)-sequencing (seq) during RANKL-induced OC differentiation and found that global chromatin accessibility decreased, especially early in OC differentiation. The global histone H3K27Ac level, an active histone modification mark, was diminished during OC differentiation by western blot and histone extract experiments. Its genomic enrichment was also reduced based on publicly available H3K27Ac chromatin immunoprecipitation (ChIP)-seq data. ATAC-seq and H3K27Ac ChIP-seq data demonstrated that RANKL induced a less accessible chromatin state during OC differentiation. Restoration of reduced H3K27Ac, presumably representing accessible states upon acetate treatment, suppresses OC differentiation by provoking immune-related gene expression. Subsequential integrative analysis of ATAC-seq, RNA-seq after acetate treatment, and H3K27Ac ChIP-seq reveals that Irf8 and its downstream targets are the most vulnerable to chromatin accessibility changes and acetate supplementation. Taken together, our study generated chromatin accessibility maps during the whole OC differentiation and suggested perturbation of chromatin accessibility might be a potential therapeutic strategy for excessive OC diseases.

3.
Int J Mol Sci ; 23(24)2022 Dec 18.
Article in English | MEDLINE | ID: mdl-36555784

ABSTRACT

Ribosomal protein S6 kinase 1 (S6K1), a key downstream effector of the mammalian target of rapamycin (mTOR), regulates diverse functions, such as cell proliferation, cell growth, and protein synthesis. Because S6K1 was previously known to be localized in the cytoplasm, its function has been mainly studied in the cytoplasm. However, the nuclear localization and function of S6K1 have recently been elucidated and other nuclear functions are expected to exist but remain elusive. Here, we show a novel nuclear role of S6K1 in regulating the expression of the Wnt target genes. Upon activation of the Wnt signaling, S6K1 translocated from the cytosol into the nucleus and subsequently bound to ß-catenin and the cofactors of the Wnt/ß-catenin transcriptional complex, leading to the upregulation of the Wnt target genes. The depletion or repression of S6K1 downregulated the Wnt target gene expression by inhibiting the formation of the Wnt/ß-catenin transcriptional complex. The S6K1-depleted colon cancer cell lines showed lower transcription levels of the Wnt/ß-catenin target genes and a decrease in the cell proliferation and invasion compared to the control cell lines. Taken together, these results indicate that nuclear S6K1 positively regulates the expression of the Wnt target genes by inducing the reciprocal interaction of the subunits of the transcriptional complex.


Subject(s)
Wnt Signaling Pathway , beta Catenin , Wnt Signaling Pathway/physiology , beta Catenin/genetics , beta Catenin/metabolism , Ribosomal Protein S6 Kinases/metabolism , Cell Nucleus/metabolism , Cell Line
4.
Exp Mol Med ; 54(2): 115-128, 2022 02.
Article in English | MEDLINE | ID: mdl-35136202

ABSTRACT

α-Synuclein is a crucial element in the pathogenesis of Parkinson's disease (PD) and related neurological diseases. Although numerous studies have presented potential mechanisms underlying its pathogenesis, the understanding of α-synuclein-mediated neurodegeneration remains far from complete. Here, we show that overexpression of α-synuclein leads to impaired DNA repair and cellular senescence. Transcriptome analysis showed that α-synuclein overexpression led to cellular senescence with activation of the p53 pathway and DNA damage responses (DDRs). Chromatin immunoprecipitation analyses using p53 and γH2AX, chromosomal markers of DNA damage, revealed that these proteins bind to promoters and regulate the expression of DDR and cellular senescence genes. Cellular marker analyses confirmed cellular senescence and the accumulation of DNA double-strand breaks. The non-homologous end joining (NHEJ) DNA repair pathway was activated in α-synuclein-overexpressing cells. However, the expression of MRE11, a key component of the DSB repair system, was reduced, suggesting that the repair pathway induction was incomplete. Neuropathological examination of α-synuclein transgenic mice showed increased levels of phospho-α-synuclein and DNA double-strand breaks, as well as markers of cellular senescence, at an early, presymptomatic stage. These results suggest that the accumulation of DNA double-strand breaks (DSBs) and cellular senescence are intermediaries of α-synuclein-induced pathogenesis in PD.


Subject(s)
Parkinson Disease , Synucleinopathies , Animals , DNA/genetics , DNA Damage , DNA Repair , Mice , Parkinson Disease/genetics , Parkinson Disease/metabolism , Tumor Suppressor Protein p53/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
5.
BMB Rep ; 54(10): 534-539, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34488930

ABSTRACT

IL-10+ regulatory B (Breg) cells play a vital role in regulating the immune responses in experimental autoimmune encephalomyelitis, colitis, and contact hypersensitivity (CHS). Several stimulants such as lipopolysaccharide (LPS), CD40 ligand, and IL-21 spur the activation and maturation of IL-10+ Breg cells, while the epigenetic mechanism for the IL-10 expression remains largely unknown. It is well accepted that the histone acetylation/ deacetylation is an important mechanism that regulates the expression of IL-10. We found that entinostat, an HDAC inhibitor, stimulated the induction of IL-10+ Breg cells by LPS in vitro and the formation of IL-10+ Breg cells to suppress CHS in vivo. We further demonstrated that entinostat inhibited HDAC1 from binding to the proximal region of the IL-10 expression promoter in splenic B cells, followed by an increase in the binding of NF-κB p65, eventually enhancing the expression of IL-10 in Breg cells. [BMB Reports 2021; 54(10): 534-539].


Subject(s)
B-Lymphocytes, Regulatory/metabolism , Benzamides/pharmacology , Dermatitis, Contact/drug therapy , Pyridines/pharmacology , Acetylation , Animals , B-Lymphocytes, Regulatory/drug effects , Benzamides/metabolism , Cells, Cultured , Colitis/metabolism , Dermatitis, Contact/genetics , Dermatitis, Contact/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Histone Deacetylase 1/drug effects , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Immunity/immunology , Immunity/physiology , Interleukin-10/immunology , Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Pyridines/metabolism , Transcription Factor RelA/metabolism
6.
Int J Mol Sci ; 22(16)2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34445596

ABSTRACT

O-linked-N-acetylglucosaminylation (O-GlcNAcylation) performed by O-GlcNAc transferase (OGT) is a nutrient-responsive post-translational modification (PTM) via the hexosamine biosynthetic pathway (HBP). Various transcription factors (TFs) are O-GlcNAcylated, affecting their activities and significantly contributing to cellular processes ranging from survival to cellular differentiation. Given the pleiotropic functions of O-GlcNAc modification, it has been studied in various fields; however, the role of O-GlcNAcylation during osteoclast differentiation remains to be explored. Kinetic transcriptome analysis during receptor activator of nuclear factor-kappaB (NF-κB) ligand (RANKL)-mediated osteoclast differentiation revealed that the nexus of major nutrient metabolism, HBP was critical for this process. We observed that the critical genes related to HBP activation, including Nagk, Gfpt1, and Ogt, were upregulated, while the global O-GlcNAcylation was increased concomitantly during osteoclast differentiation. The O-GlcNAcylation inhibition by the small-molecule inhibitor OSMI-1 reduced osteoclast differentiation in vitro and in vivo by disrupting the translocation of NF-κB p65 and nuclear factor of activated T cells c1 (NFATc1) into the nucleus by controlling their PTM O-GlcNAcylation. Furthermore, OSMI-1 had a synergistic effect with bone target therapy on osteoclastogenesis. Lastly, knocking down Ogt with shRNA (shOgt) mimicked OSMI-1's effect on osteoclastogenesis. Targeting O-GlcNAcylation during osteoclast differentiation may be a valuable therapeutic approach for osteoclast-activated bone diseases.


Subject(s)
Biosynthetic Pathways , Cell Differentiation , Hexosamines/metabolism , Osteoclasts/cytology , Protein Processing, Post-Translational , RANK Ligand/metabolism , Acylation , Animals , Cell Proliferation , Glycosylation , Male , Mice , Mice, Inbred C57BL , N-Acetylglucosaminyltransferases/metabolism , Osteoclasts/metabolism , Signal Transduction
7.
Oncogene ; 40(28): 4652-4662, 2021 07.
Article in English | MEDLINE | ID: mdl-34140644

ABSTRACT

SWItch/Sucrose Non-Fermentable (SWI/SNF) is a multiprotein complex essential for the regulation of eukaryotic gene expression. SWI/SNF complex genes are genetically altered in over 20% of human malignancies, but the aberrant regulation of the SWI/SNF subunit genes and subsequent dysfunction caused by abnormal expression of subunit gene in cancer, remain poorly understood. Among the SWI/SNF subunit genes, SMARCA4, SMARCC1, and SMARCA2 were identified to be overexpressed in human hepatocellular carcinoma (HCC). Modulation of SMARCA4, SMARCC1, and SMARCA2 inhibited in vitro tumorigenesis of HCC cells. However, SMARCA4-targeting elicited remarkable inhibition in an in vivo Ras-transgenic mouse HCC model (Ras-Tg), and high expression levels of SMARCA4 significantly associated with poor prognosis in HCC patients. Furthermore, most HCC patients (72-86%) showed SMARCA4 overexpression compared to healthy controls. To identify SMARCA4-specific active enhancers, mapping, and analysis of chromatin state in liver cancer cells were performed. Integrative analysis of SMARCA4-regulated genes and active chromatin enhancers suggested 37 genes that are strongly activated by SMARCA4 in HCC. Through chromatin immunoprecipitation-qPCR and luciferase assays, we demonstrated that SMARCA4 activates Interleukin-1 receptor-associated kinase 1 (IRAK1) expression through IRAK1 active enhancer in HCC. We then showed that transcriptional activation of IRAK1 induces oncoprotein Gankyrin and aldo-keto reductase family 1 member B10 (AKR1B10) in HCC. The regulatory mechanism of the SMARCA4-IRAK1-Gankyrin, AKR1B10 axis was further demonstrated in HCC cells and in vivo Ras-Tg mice. Our results suggest that aberrant overexpression of SMARCA4 causes SWI/SNF to promote IRAK1 enhancer to activate oncoprotein Gankyrin and AKR1B10, thereby contributing to hepatocarcinogenesis.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Oncogenes , Animals , Mice , Regulatory Sequences, Nucleic Acid
8.
Cancers (Basel) ; 13(8)2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33917757

ABSTRACT

PGC1α oppositely regulates cancer metastasis in melanoma, breast, and pancreatic cancer; however, little is known about its impact on lung cancer metastasis. Transcriptome and in vivo xenograft analysis show that a decreased PGC1α correlates with the epithelial-mesenchymal transition (EMT) and lung cancer metastasis. The deletion of a single Pgc1α allele in mice promotes bone metastasis of KrasG12D-driven lung cancer. Mechanistically, PGC1α predominantly activates ID1 expression, which interferes with TCF4-TWIST1 cooperation during EMT. Bioinformatic and clinical studies have shown that PGC1α and ID1 are downregulated in lung cancer, and correlate with a poor survival rate. Our study indicates that TCF4-TWIST1-mediated EMT, which is regulated by the PGC1α-ID1 transcriptional axis, is a potential diagnostic and therapeutic target for metastatic lung cancer.

9.
Sci Rep ; 11(1): 2942, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536477

ABSTRACT

Sepsis is caused by organ dysfunction initiated by an unrestrained host immune response to infection. The emergence of antibiotic-resistant bacteria has rapidly increased in the last decades and has stimulated a firm research platform to combat infections caused by antibiotic-resistant bacteria that cannot be eradicated with conventional antibiotics. Strategies like epigenetic regulators such as lysine demethylase (Kdm) has received attention as a new target. Thus, we sought to investigate the epigenetic mechanisms in sepsis pathophysiology with the aim of discovering new concepts for treatment. A transcriptome analysis of dendritic cells during their inflammatory state identified Kdm as a critical molecule in sepsis regulation. Next, 8-hydroxyquinoline-5-carboxylic acid (IOX1) ability to control endotoxemia induced by Lipopolysaccharide and bacterial sepsis was demonstrated. IOX1 has been shown to regulate endotoxemia and sepsis caused by Escherichia coli and carbapenem-resistant Acinetobacter baumannii and has also contributed to the suppression of multidrug-resistant bacterial growth through the inhibition of DNA Gyrase. These findings show that IOX1 could be a component agent against bacterial sepsis by functioning as a broad-spectrum antibiotic with dual effects.


Subject(s)
Acinetobacter Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Escherichia coli Infections/drug therapy , Hydroxyquinolines/pharmacology , Sepsis/drug therapy , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter baumannii/drug effects , Animals , Anti-Bacterial Agents/therapeutic use , DNA Gyrase/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli/drug effects , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Female , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Humans , Hydroxyquinolines/therapeutic use , Mice , Microbial Sensitivity Tests , Molecular Docking Simulation , Sepsis/immunology , Sepsis/microbiology
10.
Bone ; 145: 115836, 2021 04.
Article in English | MEDLINE | ID: mdl-33383217

ABSTRACT

Osteoclasts (OCs) have been well-known involved in the exacerbation of bone-related diseases. However, the role of metabolites on osteoclastogenesis has not been well characterized. Herein, we found osteoclastogenesis was negatively regulated by α-ketoglutarate (αKG) in vitro and in vivo (C57BL/6 mouse). Kinetic transcriptome analysis revealed the upregulation of solute carrier family 7 member 11 (Slc7a11), a subunit of the cysteine/glutamate antiporter, as well as the downregulation of typical OC maker genes through αKG treatment. Given that Slc7a11 could control ROS level through glutathione import, we measured intracellular ROS, then RANKL-induced ROS production was inhibited by αKG. Notably, we highlight that αKG plays an epigenetic co-factor at the Slc7a11 promoter by demethylating repressive histone H3K9 methylation and simultaneously increasing the nuclear factor erythroid 2-related factor (Nrf2) binding, a critical transcription factor through chromatin immunoprecipitation (ChIP) analysis. Together, we suggested that αKG could be a therapeutic strategy for OC activated diseases.


Subject(s)
Osteoclasts , RANK Ligand , Animals , Cell Differentiation , Epigenesis, Genetic , Glutamine , Ketoglutaric Acids , Mice , Mice, Inbred C57BL , Osteoclasts/metabolism , RANK Ligand/metabolism
11.
Cancer Res ; 81(2): 356-370, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33239431

ABSTRACT

The roles of chromatin remodelers and their underlying mechanisms of action in cancer remain unclear. In this study, SMARCB1, known initially as a bona fide tumor suppressor gene, was investigated in liver cancer. SMARCB1 was highly upregulated in patients with liver cancer and was associated with poor prognosis. Loss- and gain-of-function studies in liver cells revealed that SMARCB1 loss led to reduced cell proliferation, wound healing capacity, and tumor growth in vivo. Although upregulated SMARCB1 appeared to contribute to switch/sucrose nonfermentable (SWI/SNF) complex stability and integrity, it did not act using its known pathways antagonism with EZH2 or association between TP53 or AMPK. SMARCB1 knockdown induced a mild reduction in global H3K27 acetylation, and chromatin immunoprecipitation sequencing of SMARCB1 and acetylated histone H3K27 antibodies before and after SMARCB1 loss identified Nucleoporin210 (NUP210) as a critical target of SMARCB1, which bound its enhancer and changed H3K27Ac enrichment and downstream gene expression, particularly cholesterol homeostasis and xenobiotic metabolism. Notably, NUP210 was not only a putative tumor supporter involved in liver cancer but also acted as a key scaffold for SMARCB1 and P300 to chromatin. Furthermore, SMARCB1 deficiency conferred sensitivity to doxorubicin and P300 inhibitor in liver cancer cells. These findings provide insights into mechanisms underlying dysregulation of chromatin remodelers and show novel associations between nucleoporins and chromatin remodelers in cancer. SIGNIFICANCE: This study reveals a novel protumorigenic role for SMARCB1 and describes valuable links between nucleoporins and chromatin remodelers in cancer by identifying NUP210 as a critical coregulator of SMARCB1 chromatin remodeling activity.


Subject(s)
Gene Expression Profiling/methods , Liver Neoplasms/genetics , Nuclear Pore Complex Proteins/genetics , SMARCB1 Protein/genetics , Acetylation , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Ontology , Histones/metabolism , Humans , Kaplan-Meier Estimate , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lysine/metabolism , Nuclear Pore Complex Proteins/metabolism , SMARCB1 Protein/metabolism , Signal Transduction/genetics
12.
Int J Mol Sci ; 21(11)2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32492816

ABSTRACT

Switch/sucrose non-fermentable (SWI/SNF)-related matrix-associated actin-dependent regulator of chromatin (SMARC) subfamily B member 1 (SMARCB1) is a core subunit of the switch/sucrose non-fermentable (SWI/SNF) complex, one of the adenosine triphosphate (ATP)-dependent chromatin remodeler complexes. The unique role of SMARCB1 has been reported in various cellular contexts. Here, we focused on the general role of the ubiquitous expression of SMARCB1 in a normal cell state. We selected ARPE19 (human primary retinal pigment epithelium) and IMR90 (from human fetal lung fibroblasts) cell lines as they have completely different contexts. Furthermore, although these cell lines have been immortalized, they are relatively close to normal human cells. The loss of SMARCB1 in ARPE19 and IMR90 cells reduced cell cycle progression via the upregulation of P21. Transcriptome analysis followed by SMARCB1 knockdown in both cell lines revealed that SMARCB1 was not only involved in cell maintenance but also conferred immunomodulation. Of note, SMARCB1 bound to interleukin (IL) 6 promoter in a steady state and dissociated in an active immune response state, suggesting that SMARCB1 was a direct repressor of IL6, which was further confirmed via loss- and gain-of-function studies. Taken together, we demonstrated that SMARCB1 is a critical gatekeeper molecule of the cell cycle and immune response.


Subject(s)
Gene Expression Regulation , Retinal Pigment Epithelium/metabolism , SMARCB1 Protein/physiology , Adenosine Triphosphate/metabolism , Cell Cycle , Cell Line , Fibroblasts/metabolism , Gene Expression Profiling , Humans , Immunity , Interleukin-6/metabolism , Transcriptome
13.
Cell Death Differ ; 27(9): 2537-2551, 2020 09.
Article in English | MEDLINE | ID: mdl-32203172

ABSTRACT

E6 oncoprotein derived from high-risk human papillomavirus (HPV) drives the development of cervical cancer through p53 degradation. Because cervical cancer therapies to inactivate HPV or E6 protein are not available, alternative strategies are required. Here, we show that HPV-mediated nuclear export of human heterochromatin protein 1γ (HP1γ) reduces the stability of p53 through UBE2L3-mediated p53 polyubiquitination during cervical cancer progression. In general, HP1 plays a key role in heterochromatin formation and transcription in the nucleus. However, our immunostaining data showed that the majority of HP1γ is localized in the cytoplasm in HPV-mediated cervical cancer. We found that HPV E6 protein drives unusual nuclear export of HP1γ through the interaction between the NES sequence of HP1γ and exportin-1. The mutation of the NES sequence in HP1γ led to nuclear retention of HP1γ and reduced cervical cancer cell growth and tumor generation. We further discovered that HP1γ directly suppresses the expression of UBE2L3 which drives E6-mediated proteasomal degradation of p53 in cervical cancer. Downregulation of UBE2L3 by overexpression of HP1γ suppressed UBE2L3-dependent p53 degradation-promoting apoptosis of cervical cancer cells. Our findings propose a useful strategy to overcome p53 degradation in cervical cancer through the blockage of nuclear export of HP1γ.


Subject(s)
Carcinogenesis/pathology , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Down-Regulation/genetics , Oncogene Proteins, Viral/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Active Transport, Cell Nucleus , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Doxycycline/pharmacology , Female , Gene Expression Regulation, Neoplastic , Karyopherins/metabolism , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Protein Isoforms/metabolism , Proteolysis , Receptors, Cytoplasmic and Nuclear/metabolism , Risk Factors , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination , Exportin 1 Protein
14.
BMB Rep ; 53(2): 100-105, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31234955

ABSTRACT

While liver histopathology is heterogeneous in diabetes, the underlying mechanisms remain unclear. We investigated whether glycemic variation resulting from differential diets can induce heterogeneity in diabetic liver and the underlying molecular mechanisms. We generated end-stage non-obese diabetic model rats by subtotal-pancreatectomy in male Sprague- Dawley rats and ad libitum diet for 7 weeks (n = 33). The rats were then divided into three groups, and fed a standard- or a low-protein diet (18 or 6 kcal%, respectively), for another 7 weeks: to maintain hyperglycemia, 11 rats were fed ad libitum (18AL group); to achieve euglycemia, 11 were calorierestricted (18R group), and 11 were both calorie- and proteinrestricted with the low-protein diet (6R group). Overnightfasted liver samples were collected after the differential diets together with sham-control (18S group), and histology and molecular changes were compared. Hyperglycemic-18AL showed glycogenic hepatopathy (GH) without steatosis, with the highest GSK-3ß inactivation because of Akt activation during hyperglycemia; mitochondrial function was not impaired, compared to the 18S group. Euglycemic-18R showed neither GH nor steatosis, with intermediate GSK-3ß activation and mitochondrial dysfunction. However, euglycemic-6R showed both GH and steatosis despite the highest GSK-3ß activity and no molecular evidence of increased lipogenesis or decreased ApoB expression, where mitochondrial dysfunction was highest among the groups. In conclusion, heterogeneous liver histopathology developed in end-stage non-obese diabetic rats as the glycemic levels varied with differential diets, in which protein content in the diets as well as glycemic levels differentially influenced GSK-3ß activity and mitochondrial function in insulin-deficient state. [BMB Reports 2020; 53(2): 100-105].


Subject(s)
Diabetes Mellitus, Experimental/pathology , Glycogen Synthase Kinase 3 beta/metabolism , Hyperglycemia/pathology , Liver/pathology , Mitochondria/metabolism , Animals , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Blood Glucose/metabolism , Caloric Restriction , Diabetes Mellitus, Experimental/diet therapy , Diabetes Mellitus, Experimental/metabolism , Diet, Carbohydrate Loading , Fatty Liver/diet therapy , Fatty Liver/enzymology , Fatty Liver/metabolism , Fatty Liver/pathology , Glycemic Index/physiology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/genetics , Hepatocytes/enzymology , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Hyperglycemia/diet therapy , Hyperglycemia/enzymology , Hyperglycemia/metabolism , Insulin/metabolism , Lipogenesis , Liver/enzymology , Liver/metabolism , Male , Mitochondria/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
15.
Cell Death Differ ; 27(6): 1844-1861, 2020 06.
Article in English | MEDLINE | ID: mdl-31819156

ABSTRACT

Long noncoding RNAs (lncRNAs) regulating diverse cellular processes implicate in many diseases. However, the function of lncRNAs in cellular senescence remains largely unknown. Here we identify a novel long intergenic noncoding RNA Linc-ASEN expresses in prematurely senescent cells. We find that Linc-ASEN associates with UPF1 by RNA pulldown mass spectrometry analysis, and represses cellular senescence by reducing p21 production transcriptionally and posttranscriptionally. Mechanistically, the Linc-ASEN-UPF1 complex suppressed p21 transcription by recruiting Polycomb Repressive Complex 1 (PRC1) and PRC2 to the p21 locus, and thereby preventing binding of the transcriptional activator p53 on the p21 promoter through histone modification. In addition, the Linc-ASEN-UPF1 complex repressed p21 expression posttranscriptionally by enhancing p21 mRNA decay in association with DCP1A. Accordingly, Linc-ASEN levels were found to correlate inversely with p21 mRNA levels in tumors from patient-derived mouse xenograft, in various human cancer tissues, and in aged mice tissues. Our results reveal that Linc-ASEN prevents cellular senescence by reducing the transcription and stability of p21 mRNA in concert with UPF1, and suggest that Linc-ASEN might be a potential therapeutic target in processes influenced by senescence, including cancer.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Neoplasms/metabolism , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/metabolism , RNA, Long Noncoding/metabolism , Animals , Cell Line, Tumor , Cellular Senescence , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude
16.
Int Immunopharmacol ; 73: 482-490, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31173970

ABSTRACT

There is currently no specific drug for the treatment of sepsis and antibiotic administration is considered the best option, despite numerous issues. Therefore, the development of drugs to control the pathogen-induced inflammatory responses associated with sepsis is essential. To address this, our study examined the transcriptomes of lipopolysaccharide (LPS)-induced dendritic cells (DCs), identifying TANK-binding kinase1 (Tbk1) as a key factor involved in the inflammatory response. These data suggested drug repositioning of the Tbk1 inhibitor CYT387, currently used for the treatment of myelofibrosis and some cancers, as a candidate for regulating the LPS-induced inflammatory response. CYT387 also inhibited pro-inflammatory cytokine and surface molecule expression by mature DCs after LPS exposure. These effects correlated with both Akt phosphorylation and IκBα degradation. Finally, CYT387 demonstrated therapeutic effects in LPS-induced endotoxemia and Escherichia coli K1-induced mouse models of sepsis and decreased the expression of pro-inflammatory cytokines. In conclusion, our study suggests that drug repositioning of CYT387 may serve as a potential therapeutic for sepsis.


Subject(s)
Benzamides/therapeutic use , Endotoxemia/drug therapy , Escherichia coli Infections/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/therapeutic use , Sepsis/drug therapy , Animals , Benzamides/pharmacology , Cytokines/immunology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Drug Repositioning , Endotoxemia/immunology , Escherichia coli Infections/immunology , Female , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Sepsis/immunology , Transcriptome/drug effects
17.
Biochem Biophys Res Commun ; 511(4): 746-752, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30833074

ABSTRACT

SOX9 is a key transcription factor during cell differentiation, sex determination, and tumorigenesis. However, the detailed mechanisms of its targeting strategy remain elusive. To investigate possibilities of targeting SOX9 with epigenetic drugs and the precise underlying mechanisms, two human cancer cell lines were chosen as model systems, which showed high SOX9 expression and anti-tumorigenic effects upon loss of SOX9. Histone acetylation-related screening of a small panel of epigenetic drugs revealed that the bromodomain reader inhibitor JQ1 dramatically downregulated SOX9 through multiple regulation steps, namely, transcription, BRD4-SOX9 protein-protein interaction, and further protein stability. These findings suggest that BRD4 inhibition is a novel therapeutic strategy for diseases characterized by SOX9 overexpression.


Subject(s)
Azepines/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Down-Regulation/drug effects , SOX9 Transcription Factor/genetics , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Cell Line, Tumor , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/genetics
18.
Gut ; 68(4): 708-720, 2019 04.
Article in English | MEDLINE | ID: mdl-29475852

ABSTRACT

OBJECTIVE: Alcoholic liver disease (ALD) is a leading cause of death among chronic liver diseases. However, its pathogenesis has not been completely established. MicroRNAs (miRNAs) are key contributors to liver diseases progression. This study investigated hepatocyte-abundant miRNAs dysregulated by ALD, its impact on hepatocyte injury and the underlying basis. DESIGN: Alcoholic hepatitis (AH) human and animal liver samples and hepatocytes were used to assess miR-148a levels. Pre-miR-148a was delivered specifically to hepatocytes in vivo using lentivirus. Immunoblottings, luciferase reporter assays, chromatin immunoprecipitation and immunofluorescence assays were carried out in cell models. RESULTS: The miRNA profile and PCR analyses enabled us to find substantial decrease of miR-148a in the liver of patients with AH. In mice subjected to Lieber-DeCarli alcohol diet or binge alcohol drinking, miR-148a levels were also markedly reduced. In cultured hepatocytes and mouse livers, alcohol exposure inhibited forkhead box protein O1 (FoxO1) expression, which correlated with miR-148a levels and significantly decreased in human AH specimens. FoxO1 was identified as a transcription factor for MIR148A transactivation. MiR-148a directly inhibited thioredoxin-interacting protein (TXNIP) expression. Consequently, treatment of hepatocytes with ethanol resulted in TXNIP overexpression, activating NLRP3 inflammasome and caspase-1-mediated pyroptosis. These events were reversed by miR-148a mimic or TXNIP small-interfering RNA transfection. Hepatocyte-specific delivery of miR-148a to mice abrogated alcohol-induced TXNIP overexpression and inflammasome activation, attenuating liver injury. CONCLUSION: Alcohol decreases miR-148a expression in hepatocytes through FoxO1, facilitating TXNIP overexpression and NLRP3 inflammasome activation, which induces hepatocyte pyroptosis. Our findings provide information on novel targets for reducing incidence and progression of ALD.


Subject(s)
Carrier Proteins/metabolism , Hepatitis, Alcoholic/metabolism , Hepatocytes/metabolism , Inflammasomes/metabolism , Pyroptosis , Thioredoxins/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Disease Progression , Fluorescent Antibody Technique , Humans , Immunoblotting , Mice , MicroRNAs , Polymerase Chain Reaction
19.
Int J Oncol ; 53(1): 395-403, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29749437

ABSTRACT

The interleukin-7 receptor (IL7R) is generally expressed in immune cells and is critical in survival, development and homeostasis in the immune system. Advanced genome-wide cancer studies have reported that IL7R is genetically amplified in human esophageal squamous cell carcinoma (ESCC), however, the exact role of IL7R in ESCC has not been investigated. In the present study, it was found that IL7R was overexpressed in ESCC cohorts and the loss of IL7R induced anti-oncogenic effects in ESCC cell lines. A small panel of epigenetic drugs were screened for their ability to downregulate the expression of IL7R. Unexpectedly, apicidin, a histone deacetylase (HDAC) inhibitor, effectively downregulated the expression of IL7R in a dose-dependent manner at an early time-point, as determined by quantitative polymerase chain reaction and IL7R immunostaining, and did not require de novo protein synthesis. Of note, apicidin induced the acetylation of Forkhead box-containing protein, O subfamily 1, which acts as a repressor at the IL7R promoter, accompanied with depleted active histone modifications based on chromatin immunoprecipitation assay. Taken together, these results demonstrated that targeting oncogenic IL7R in ESCC by HDAC inhibitors may be a valuable therapeutic approach.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Esophageal Neoplasms/drug therapy , Forkhead Box Protein O1/genetics , Histone Deacetylase Inhibitors/pharmacology , Interleukin-7 Receptor alpha Subunit/genetics , Acetylation/drug effects , Apoptosis/drug effects , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Gene Expression Regulation, Neoplastic/drug effects , Humans , Peptides, Cyclic/pharmacology
20.
Gut ; 67(1): 166-178, 2018 01.
Article in English | MEDLINE | ID: mdl-28341749

ABSTRACT

OBJECTIVE: Interferons (IFNs) mediate direct antiviral activity. They play a crucial role in the early host immune response against viral infections. However, IFN therapy for HBV infection is less effective than for other viral infections. DESIGN: We explored the cellular targets of HBV in response to IFNs using proteome-wide screening. RESULTS: Using LC-MS/MS, we identified proteins downregulated and upregulated by IFN treatment in HBV X protein (HBx)-stable and control cells. We found several IFN-stimulated genes downregulated by HBx, including TRIM22, which is known as an antiretroviral protein. We demonstrated that HBx suppresses the transcription of TRIM22 through a single CpG methylation in its 5'-UTR, which further reduces the IFN regulatory factor-1 binding affinity, thereby suppressing the IFN-stimulated induction of TRIM22. CONCLUSIONS: We verified our findings using a mouse model, primary human hepatocytes and human liver tissues. Our data elucidate a mechanism by which HBV evades the host innate immune system.


Subject(s)
5' Untranslated Regions/genetics , CpG Islands/genetics , Hepatitis B virus/immunology , Interferons/immunology , Minor Histocompatibility Antigens/genetics , Repressor Proteins/genetics , Tripartite Motif Proteins/genetics , Animals , Down-Regulation/genetics , Down-Regulation/immunology , Epigenesis, Genetic , Gene Expression Regulation/immunology , Hepatocytes/metabolism , Humans , Immune Evasion , Liver/metabolism , Methylation , Mice , Minor Histocompatibility Antigens/biosynthesis , Proteome , Repressor Proteins/biosynthesis , Tripartite Motif Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...