Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Genomics ; 114(1): 1-8, 2022 01.
Article in English | MEDLINE | ID: mdl-34822968

ABSTRACT

Diurnal oscillations in gene expression are a hallmark of the liver internal clock and can be regulated by a variety of environmental stimuli. The circadian rhythm and liver regeneration (LR) are intimately linked. However, how they affect each other at the transcriptomic level is mainly unknown. Here, we revealed that partial hepatectomy (PHx)-induced LR led to reprogramming of rhythmic gene expression profiles as a consequence of disrupted BMAL1 occupation on the chromatin, while the rhythm of core clock genes remained robust. Furthermore, we demonstrated retarded LR when PHx was carried out in the evening, possibly due to the accumulation of DEC1. In summary, our data offer a broad perspective of the relationship between circadian rhythm and LR and suggest that the timing of PHx should be considered in the clinic application.


Subject(s)
Circadian Rhythm , Liver , Circadian Rhythm/genetics , Liver/metabolism , Transcriptome
2.
Biochem Biophys Res Commun ; 606: 75-79, 2022 05 28.
Article in English | MEDLINE | ID: mdl-35339755

ABSTRACT

Hydroxyacyl-CoA dehydrogenase (HADH) catalyzes the third reaction of mitochondrial ß-oxidation cascade, while the regulation of its expression and function remains to be elucidated. Using the quantitative translation initiation sequencing (QTI-seq), we have identified that murine Hadh mRNA has two alternative translation start codons. We demonstrated that translation from upstream start codon encodes the mitochondrial isoform of HADH, while translation from downstream start codon produces a short isoform (HADH-S) with predominant nuclear localization. Moreover, overexpression of HADH-S inhibits the proliferation of mouse embryonic fibroblasts. Overall, our results identify a novel isoform of HADH participating in cell proliferation.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases , Fibroblasts , 3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Animals , Cell Proliferation , Codon, Initiator , Fibroblasts/metabolism , Mice , Protein Isoforms/genetics
3.
EMBO Rep ; 21(4): e49229, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32064749

ABSTRACT

N6-methyladenosine (m6 A) mRNA methylation has emerged as an important player in many biological processes by regulating gene expression. However, its roles in intestinal stem cell (ISC) homeostasis remain largely unknown. Here, we report that YTHDF1, an m6 A reader, is highly expressed in ISCs and its expression is upregulated by Wnt signaling at the translational level. Whereas YTHDF1 is dispensable for normal intestinal development in mice, genetic ablation of Ythdf1 dramatically blocks Wnt-driven regeneration and tumorigenesis with reduced ISC stemness. Mechanistically, YTHDF1 facilitates the translation of Wnt signaling effectors including TCF7L2/TCF4, while this process is enhanced during Wnt activation to augment ß-catenin activity. Targeting YTHDF1 in ISCs of established tumors leads to tumor shrinkage and prolonged survival. Collectively, our studies unveil YTHDF1 as an amplifier of Wnt/ß-catenin signaling at the translational level, which is required for the maintenance of ISCs during regeneration and tumorigenesis.


Subject(s)
Intestines , Wnt Signaling Pathway , Animals , Carcinogenesis , Cell Transformation, Neoplastic , Methylation , Mice
4.
Biochem Biophys Res Commun ; 557: 85-89, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33862464

ABSTRACT

N6-methyladenosine (m6A) mRNA modification has been defined as a crucial regulator in various biological processes. Recent studies indicated an essential role of YTHDF1, an m6A reader, in the maintenance of intestinal stem cells (ISCs), while the detailed mechanism remains to be explored. By searching our m6A sequencing, RNA sequencing, and ribosome profiling data, we identified the transcriptional enhanced associate domain 1 (TEAD1) as a direct target of YTHDF1. We confirmed the presence of m6A modifications in TEAD1 mRNA and its binding with YTHDF1. Knockdown of either m6A methyltransferase METTL3 or YTHDF1 reduced the translation of TEAD1. TEAD1 was highly expressed in ISCs, while depletion of TEAD1 inhibited proliferation and induced differentiation of organoids. Overexpression of TEAD1 reversed the impaired stemness elicited by YTHDF1 depletion. These findings identify TEAD1 as a functional target of m6A-YTHDF1 in sustaining intestinal stemness.


Subject(s)
DNA-Binding Proteins/biosynthesis , Intestines/cytology , Nuclear Proteins/biosynthesis , RNA-Binding Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/biosynthesis , Adenosine/analogs & derivatives , Adenosine/genetics , Adenosine/metabolism , Animals , Cell Line , Cell Proliferation/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HCT116 Cells , Humans , Intestines/physiology , Methylation , Mice , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organoids , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , TEA Domain Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Cell Mol Life Sci ; 77(15): 2887-2898, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32060582

ABSTRACT

Liver regeneration (LR) capacity in vertebrates developed through natural selection over a hundred million years of evolution. To maintain homeostasis or recover from various injuries, liver cells must regenerate; this process includes the renewal of parenchymal and nonparenchymal cells as well as the formation of liver structures. The cellular origin of newly grown tissue is one of the critical questions in this area and has been a subject of prolonged debate. The regenerative tissue may derive from either hepatocyte self-duplication or liver stem/progenitor cells (LSPCs). Recently, hepatocyte subpopulations and cholangiocytes were also described as important founder cells. The niche that triggers the proliferation of hepatocytes and the differentiation of LSPCs has been extensively studied. Meanwhile, in vitro culture systems for liver founder cells and organoids have been developed rapidly for mechanistic studies and potential therapeutic purposes. This review summarizes the cellular sources and niches that give rise to renewed hepatocytes during LR, and it also describes in vitro culture studies of those founder cells for future applications, as well as current reports for stem cell-based therapies for liver diseases.


Subject(s)
Hepatocytes/cytology , Liver Regeneration/physiology , Animals , Cell Differentiation , Cell- and Tissue-Based Therapy , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/therapy , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/transplantation , Hepatocytes/metabolism , Hepatocytes/transplantation , Humans , Stem Cells/cytology , Stem Cells/metabolism
6.
J Biol Chem ; 289(13): 8947-59, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24554706

ABSTRACT

Mutant p53 proteins (mutp53) often acquire oncogenic activities, conferring drug resistance and/or promoting cancer cell migration and invasion. Although it has been well established that such a gain of function is mainly achieved through interaction with transcriptional regulators, thereby modulating cancer-associated gene expression, how the mutp53 function is regulated remains elusive. Here we report that activating transcription factor 3 (ATF3) bound common mutp53 (e.g. R175H and R273H) and, subsequently, suppressed their oncogenic activities. ATF3 repressed mutp53-induced NFKB2 expression and sensitized R175H-expressing cancer cells to cisplatin and etoposide treatments. Moreover, ATF3 appeared to suppress R175H- and R273H-mediated cancer cell migration and invasion as a consequence of preventing the transcription factor p63 from inactivation by mutp53. Accordingly, ATF3 promoted the expression of the metastasis suppressor SHARP1 in mutp53-expressing cells. An ATF3 mutant devoid of the mutp53-binding domain failed to disrupt the mutp53-p63 binding and, thus, lost the activity to suppress mutp53-mediated migration, suggesting that ATF3 binds to mutp53 to suppress its oncogenic function. In line with these results, we found that down-regulation of ATF3 expression correlated with lymph node metastasis in TP53-mutated human lung cancer. We conclude that ATF3 can suppress mutp53 oncogenic function, thereby contributing to tumor suppression in TP53-mutated cancer.


Subject(s)
Activating Transcription Factor 3/metabolism , Carcinogenesis/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Cell Movement/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , NF-kappa B p52 Subunit/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
7.
Diabetes ; 72(8): 1083-1094, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37224383

ABSTRACT

The induction of beige adipocytes in white adipose tissue (WAT), also known as WAT beiging, improves glucose and lipid metabolism. However, the regulation of WAT beiging at the posttranscriptional level remains to be studied. Here, we report that METTL3, the methyltransferase of N6-methyladenosine (m6A) mRNA modification, is induced during WAT beiging in mice. Adipose-specific depletion of the Mettl3 gene undermines WAT beiging and impairs the metabolic capability of mice fed with a high-fat diet. Mechanistically, METTL3-catalyzed m6A installation on thermogenic mRNAs, including Krüppel-like factor 9 (Klf9), prevents their degradation. Activation of the METTL3 complex by its chemical ligand methyl piperidine-3-carboxylate promotes WAT beiging, reduces body weight, and corrects metabolic disorders in diet-induced obese mice. These findings uncover a novel epitranscriptional mechanism in WAT beiging and identify METTL3 as a potential therapeutic target for obesity-associated diseases. ARTICLE HIGHLIGHTS: METTL3, the methyltransferase of N6-methyladenosine (m6A) mRNA modification, is induced during WAT beiging. Depletion of Mettl3 undermines WAT beiging and impairs thermogenesis. METTL3-mediated m6A installation promotes the stability of Krüppel-like factor 9 (Klf9). KLF9 rescues impaired beiging elicited by Mettl3 depletion. Pharmaceutical activation of the METTL3 complex by its chemical ligand methyl piperidine-3-carboxylate induces WAT beiging. Methyl piperidine-3-carboxylate corrects obesity-associated disorders. The METTL3-KLF9 pathway may serve as a potential therapeutic target for obesity-associated diseases.


Subject(s)
Adipose Tissue, White , Obesity , Animals , Mice , Adipose Tissue, White/metabolism , Kruppel-Like Transcription Factors/metabolism , Ligands , Methyltransferases/genetics , Methyltransferases/metabolism , Obesity/genetics , Obesity/metabolism , Piperidines , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Nat Commun ; 14(1): 1379, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36914671

ABSTRACT

Obesity, one of the most serious public health issues, is caused by the imbalance of energy intake and energy expenditure. N(6)-methyladenosine (m6A) RNA modification has been recently identified as a key regulator of obesity, while the detailed mechanism is elusive. Here, we find that YTH RNA binding protein 1 (YTHDF1), an m6A reader, acts as an essential regulator of white adipose tissue metabolism. The expression of YTHDF1 decreases in adipose tissue of male mice fed a high-fat diet. Adipocyte-specific Ythdf1 deficiency exacerbates obesity-induced metabolic defects and inhibits beiging of inguinal white adipose tissue (iWAT) in male mice. By contrast, male mice with WAT-specific YTHDF1 overexpression are resistant to obesity and shows promotion of beiging. Mechanistically, YTHDF1 regulates the translation of diverse m6A-modified mRNAs. In particular, YTHDF1 facilitates the translation of bone morphogenetic protein 8b (Bmp8b) in an m6A-dependent manner to induce the beiging process. Here, we show that YTHDF1 may be an potential therapeutic target for the management of obesity-associated diseases.


Subject(s)
Adipocytes , Adipose Tissue, White , RNA-Binding Proteins , Animals , Male , Mice , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism , Obesity/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
9.
J Biol Chem ; 285(47): 36857-64, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20843798

ABSTRACT

Solid tumor development is frequently accompanied by energy-deficient conditions such as glucose deprivation and hypoxia. Follistatin (FST), a secretory protein originally identified from ovarian follicular fluid, has been suggested to be involved in tumor development. However, whether it plays a role in cancer cell survival under energy-deprived conditions remains elusive. In this study, we demonstrated that glucose deprivation markedly enhanced the expression and nucleolar localization of FST in HeLa cells. The nucleolar localization of FST relied on its nuclear localization signal (NLS) comprising the residues 64-87. Localization of FST to the nucleolus attenuated rRNA synthesis, a key process for cellular energy homeostasis and cell survival. Overexpression of FST delayed glucose deprivation-induced apoptosis, whereas down-regulation of FST exerted the opposite effect. These functions depended on the presence of an intact NLS because the NLS-deleted mutant of FST lost the rRNA inhibition effect and the cell protective effect. Altogether, we identified a novel nucleolar function of FST, which is of importance in the modulation of cancer cell survival in response to glucose deprivation.


Subject(s)
Apoptosis , Cell Nucleolus/metabolism , Follistatin/metabolism , Glucose/deficiency , RNA, Ribosomal/biosynthesis , Uterine Cervical Neoplasms/pathology , Blotting, Northern , Blotting, Western , Chromatin Immunoprecipitation , Down-Regulation , Female , Fluorescent Antibody Technique , Follistatin/antagonists & inhibitors , Follistatin/genetics , HeLa Cells , Humans , Nuclear Localization Signals , RNA, Messenger/genetics , RNA, Ribosomal/antagonists & inhibitors , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Uterine Cervical Neoplasms/metabolism
10.
Cancer Lett ; 518: 256-265, 2021 10 10.
Article in English | MEDLINE | ID: mdl-34339799

ABSTRACT

N6-methyladenosine (m6A), the most prevalent internal modification in eukaryotic mRNAs, regulates gene expression at the post-transcriptional level. The reader proteins of m6A, mainly YTH domain-containing proteins, specifically recognize m6A-modified mRNAs and regulate their metabolism. Recent studies have highlighted essential roles of m6A readers in the initiation and development of human cancers. In this review, we summarize recent findings about the biological functions of YTH domain proteins in cancers, the underlying mechanisms, and clinical implications. Gene expression reprogramming by dysregulated m6A reader proteins offers potential targets for cancer treatment, while targeted m6A editors and readers provide tools to manipulate m6A metabolism in cancers.


Subject(s)
Adenosine/analogs & derivatives , Neoplasms/genetics , Protein Domains/genetics , RNA, Messenger/genetics , Adenosine/genetics , Animals , Gene Expression/genetics , Humans , Methylation , RNA Processing, Post-Transcriptional/genetics
11.
Aging (Albany NY) ; 13(19): 23119-23132, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34620733

ABSTRACT

The anticancer effect of zinc oxide nanoparticles (ZnO NPs) largely relies on cellular responses such as alteration of gene expression. Although ZnO NPs have been reported to induce transcriptional changes, the potential of ZnO NPs to affect cellular translatome remains largely unknown. Using ribosome profiling, we demonstrated that the transcription of 78 genes and the translation of 1,448 genes are affected during one hour of ZnO NPs exposure in A549 human lung cancer cells. The mitogen-activated protein kinase (MAPK) pathway is up-regulated upon ZnO NP treatment. The upstream open reading frame (uORF) plays a pervasive role in the induction of up-regulated genes, including TLNRD1 and CCNB1IP1. Knockdown of TLNRD1 or CCNB1IP1 reduces ZnO NP-induced cytotoxicity. Together, our study characterizes the landscape of translational alteration under ZnO NPs treatment and provides potential targets to augment the anticancer effect of ZnO NPs.


Subject(s)
Metal Nanoparticles/chemistry , Ribosomes/drug effects , Sequence Analysis, RNA/methods , Zinc Oxide/pharmacology , A549 Cells , Genetic Structures , Humans , Protein Biosynthesis , RNA, Messenger/genetics , Ribosomes/genetics , Zinc Oxide/chemistry
12.
J Exp Clin Cancer Res ; 38(1): 217, 2019 May 23.
Article in English | MEDLINE | ID: mdl-31122284

ABSTRACT

BACKGROUND: Upregulation of RNA polymerase (Pol) III products, including tRNAs and 5S rRNA, in tumor cells leads to enhanced protein synthesis and tumor formation, making it a potential target for cancer treatment. In this study, we evaluated the inhibition of Pol III transcription by triptolide and the anti-cancer effect of this drug in colorectal tumorigenesis. METHODS: The effect of triptolide on colorectal cancer development was assessed in colorectal cancer mouse models, 3D organoids, and cultured cells. Colorectal cancer cells were treated with triptolide. Pol III transcription was measured by real-time quantitative polymerase chain reaction (PCR). The formation of TFIIIB, a multi-subunit transcription factor for Pol III, was determined by chromatin immunoprecipitation (ChIP), co-immunoprecipitation (Co-IP), and fluorescence resonance energy transfer (FRET). RESULTS: Triptolide reduced both tumor number and tumor size in adenomatous polyposis coli (Apc) mutated (ApcMin/+) mice as well as AOM/DSS-induced mice. Moreover, triptolide effectively inhibited colorectal cancer cell proliferation, colony formation, and organoid growth in vitro, which was associated with decreased Pol III target genes. Mechanistically, triptolide treatment blocked TBP/Brf1interaction, leading to the reduced formation of TFIIIB at the promoters of tRNAs and 5S rRNA. CONCLUSIONS: Together, our data suggest that inhibition of Pol III transcription with existing drugs such as triptolide provides a new avenue for developing novel therapies for colorectal cancer.


Subject(s)
Colorectal Neoplasms/drug therapy , Diterpenes/administration & dosage , Phenanthrenes/administration & dosage , Transcription Factor TFIIIB/metabolism , Transcription, Genetic/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Diterpenes/pharmacology , Epoxy Compounds/administration & dosage , Epoxy Compounds/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Mice , Phenanthrenes/pharmacology , Promoter Regions, Genetic , RNA, Ribosomal, 5S , RNA, Transfer/genetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Chin Med J (Engl) ; 132(3): 311-318, 2019 Feb 05.
Article in English | MEDLINE | ID: mdl-30681497

ABSTRACT

BACKGROUND: The clinical trials emerged centromere protein E inhibitor GSK923295 as a promising anticancer drug, but its function in hepatocellular carcinoma (HCC) remain needs to be fully elucidated, especially as chemotherapy after hepatectomy for liver tumors. We aimed to describe anti-HCC activities of GSK923295 and compare its antiproliferative effects on liver regeneration after partial hepatectomy (PH). METHODS: All subjects were randomized to treatment with either vehicle or GSK923295. Antitumor activity of GSK923295 was assessed by xenograft growth assays. The C57BL/6 mice were subjected to 70% PH and the proliferation was calculated by liver coefficient, further confirmed by immunohistochemistry. The proliferation and cell cycle analysis of liver cell AML12 and HCC cells LM3, HUH7, and HepG2 were investigated using the cell counting kit-8 assay and Flow Cytometry. The chromosome misalignment and segregation in AML12 cells were visualized by immunofluorescence. RESULTS: Treatment with GSK923295 induced antiproliferation in HCC cell lines. It also caused delay on HCC tumor growth instead of regression both in a HCC cell line xenograft model and patient-derived tumor xenograft model. With microarray analysis, CENtromere Protein E was gradually increased in mouse liver after PH. Exposure of liver cells to GSK923295 resulted in delay on a cell cycle in mitosis with a phenotype of misaligned chromosomes and chromosomes clustered. In 70% PH mouse model, GSK923295 treatment also remarkably reduced liver regeneration in later stage, in parallel with the mitotic marker phospho-histone H3 elevation. CONCLUSION: The anticancer drug GSK923295 causes a significant delay on HCC tumor growth and liver regeneration after PH in later stage.


Subject(s)
Antineoplastic Agents/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/surgery , Liver Neoplasms/drug therapy , Liver Neoplasms/surgery , Sarcosine/analogs & derivatives , Animals , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Electrophoresis, Polyacrylamide Gel , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Liver Regeneration/physiology , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Sarcosine/therapeutic use , Xenograft Model Antitumor Assays
14.
Talanta ; 194: 183-188, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30609520

ABSTRACT

Micelle-hybridized supramolecular hydrogels were constructed through the self-assembly of gelator N,N-dibenzoyl-L-cystine (DBC) and micelles formed from a Gemini surfactant (G12-8-12). A phosphor, palladium meso-tetra (4-carboxyphenyl) porphyrin (Pd-TCPP) and Al3+ ions were loaded within the hybrid system. Interestingly, the room temperature phosphorescence (RTP) of Pd-TCPP can be efficiently enhanced and modulated by the concentration of Al3+ ions. The enhancement effect could be attributed to the interactions between Al3+ and DBC as well as porphyrin, which verified by 1H NMR analysis. The study of transmission electron microscopy and scanning electron microscopy indicated that a more compact 3D network structure of the gel system was formed upon the addition of Al3+. In addition, measurement of critical micelle concentration indicated that Al3+ ions increase the surface activity of G12-8-12 to promote micelle formation, thereby increasing the dispersion of Pd-TCPP in the hybrid gels. Based on the synergistic effect of these results, the non-radiative transition of Pd-TCPP was efficiently inhibited, resulting in highly efficient RTP. Furthermore, the enhanced RTP of as-prepared gel system shows potential application to detect trace Hg2+ ions because the RTP can be quenched by Hg2+. A linear relationship between RTP against the logarithmic concentration of Hg2+ was found over the range of 6 × 10-8 and 1 × 10-6 mol/L. The detection limit was found to be 0.017 nmol/L.

15.
Int J Biochem Cell Biol ; 113: 95-102, 2019 08.
Article in English | MEDLINE | ID: mdl-31220582

ABSTRACT

ZnO nanoparticle (ZnO NP) exposure causes oxidative stress in the respiratory system, leading to pulmonary damage. Activating transcription factor 3 (ATF3) participates in a variety of cellular stress responses. However, the role of ATF3 in ZnO NP genotoxicity and cytotoxicity remains to be explored. Here we reported that ZnO NP treatment dramatically induced the expression of ATF3 in human bronchial epithelial (HBE) cells, which was mediated by the nuclear factor erythroid 2-related factor 2 (Nrf2). ATF3 was required for the repair of ZnO NP-induced DNA damage as gamma foci number increased when endogenous ATF3 was silenced. Moreover, ATF3 also contributed to ZnO NP-induced cell apoptosis. Mechanistic study revealed that ATF3 interacted with the p53 protein and upregulated its expression under ZnO NP treatment. Collectively, our findings demonstrated ATF3 as an important regulator of epithelial homeostasis by promoting both DNA repair and the death of damaged cells under ZnO NP-induced genotoxic stress.


Subject(s)
Activating Transcription Factor 3/metabolism , Bronchi/drug effects , Epithelial Cells/drug effects , Nanoparticles/toxicity , Zinc Oxide/toxicity , Activating Transcription Factor 3/genetics , Apoptosis/drug effects , Bronchi/metabolism , Bronchi/pathology , Cells, Cultured , DNA Damage , DNA Repair , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Mutagens/chemistry , Mutagens/toxicity , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Nanoparticles/chemistry , Transcription, Genetic/drug effects , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Zinc Oxide/chemistry
16.
Cell Death Dis ; 10(2): 127, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30755584

ABSTRACT

Silica nanoparticles (SiNPs) have been reported to induce pulmonary fibrosis (PF) with an unknown mechanism. Recently, the activation of autophagy, a lysosome-dependent cell degradation pathway, by SiNPs has been identified in alveolar epithelial cells (AECs). However, the underlying mechanism and the relevance of SiNPs-induced autophagy to the development of PF remain elusive. Here, we report that autophagy dysfunction and subsequent apoptosis in AECs are involved in SiNPs-induced PF. SiNPs engulfed by AECs enhance autophagosome accumulation and apoptosis both in vivo and in vitro. Mechanically, SiNPs block autophagy flux through impairing lysosomal degradation via acidification inhibition. Lysosomal reacidification by cyclic-3',5'-adenosine monophosphate (cAMP) significantly enhances autophagic degradation and attenuate apoptosis. Importantly, enhancement of autophagic degradation by rapamycin protects AECs from apoptosis and attenuates SiNPs-induced PF in the mouse model. Altogether, our data demonstrate a repressive effect of SiNPs on lysosomal acidification, contributing to the decreased autophagic degradation in AECs, thus leading to apoptosis and subsequent PF. These findings may provide an improved understanding of SiNPs-induced PF and molecular targets to antagonize it.


Subject(s)
Alveolar Epithelial Cells/metabolism , Autophagy/drug effects , Nanoparticles/administration & dosage , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Silicon Dioxide/pharmacology , A549 Cells , Animals , Apoptosis/drug effects , Cell Survival , Cyclic AMP/metabolism , Cyclic AMP/pharmacology , Disease Models, Animal , Endosomes/metabolism , Humans , Hydrogen-Ion Concentration/drug effects , Lysosomes/metabolism , Male , Mice , Nanoparticles/adverse effects , Nanoparticles/chemistry , Signal Transduction/drug effects , Silicon Dioxide/administration & dosage , Silicon Dioxide/adverse effects , Silicon Dioxide/chemistry , Transfection
17.
Sci Rep ; 6: 21133, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26878911

ABSTRACT

Silica nanoparticles (SiO2 NPs) cause oxidative stress in respiratory system. Meanwhile, human cells launch adaptive responses to overcome SiO2 NP toxicity. However, besides a few examples, the regulation of SiO2 NP-responsive proteins and their functions in SiO2 NP response remain largely unknown. In this study, we demonstrated that SiO2 NP induced the expression of follistatin (FST), a stress responsive gene, in mouse lung tissue as well as in human lung epithelial cells (A549). The levels of Ac-H3(K9/18) and H3K4me2, two active gene markers, at FST promoter region were significantly increased during SiO2 NP treatment. The induction of FST transcription was mediated by the nuclear factor erythroid 2-related factor 2 (Nrf2), as evidenced by the decreased FST expression in Nrf2-deficient cells and the direct binding of Nrf2 to FST promoter region. Down-regulation of FST promoted SiO2 NP-induced apoptosis both in cultured cells and in mouse lung tissue. Furthermore, knockdown of FST increased while overexpression of FST decreased the expression level of NADPH oxidase 1 (NOX1) and NOX5 as well as the production of cellular reactive oxygen species (ROS). Taken together, these findings demonstrated a protective role of FST in SiO2 NP-induced oxidative stress and shed light on the interaction between SiO2 NPs and biological systems.


Subject(s)
Alveolar Epithelial Cells/metabolism , Follistatin/genetics , NF-E2-Related Factor 2/metabolism , Nanoparticles , Oxidative Stress , Silicon Dioxide , Transcriptional Activation , Animals , Apoptosis/genetics , Cell Line , Gene Expression , Humans , Male , Membrane Proteins/genetics , Mice , NADPH Oxidase 1 , NADPH Oxidase 5 , NADPH Oxidases/genetics , Nanoparticles/adverse effects , Reactive Oxygen Species/metabolism , Silicon Dioxide/adverse effects
18.
PLoS One ; 8(8): e71487, 2013.
Article in English | MEDLINE | ID: mdl-23977052

ABSTRACT

Nuclear translocation of angiogenin (ANG) is essential for the proliferation of its target cells. ANG promotes rRNA synthesis, while whether it regulates mRNA transcription remains unknown. Using the chromatin immunoprecipitation method, we have identified 12 ANG-binding sequences. One of these sequences lies in the estrogen receptor-related receptor gamma (ERRγ) gene which we designated as ANG-Binding Sequence within ERRγ (ABSE). ABSE exhibited ANG-dependent repressor activity in the luciferase reporter system. Down-regulation of ANG increased ERRγ expression, and active gene marker level at the ABSE region. The expression levels of ERRγ targets genes, p21(WAF/CIP) and p27(KIP1), and the occupation of ERRγ on their promoter regions were increased in ANG-deficient cells accordingly. Furthermore, knockdown of ERRγ promoted the proliferation rate in ANG-deficient breast cancer cells. Finally, immunohistochemistry staining showed negative correlation between ANG and ERRγ in breast cancer tissue. Altogether, our study provides evidence that nuclear ANG directly binds to the ABSE of ERRγ gene and inhibits ERRγ transcription to promote breast cancer cell proliferation.


Subject(s)
Receptors, Estrogen/metabolism , Ribonuclease, Pancreatic/metabolism , Transcription, Genetic , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Immunohistochemistry , Male , Protein Binding/genetics , Protein Processing, Post-Translational , RNA Polymerase II/metabolism
19.
PLoS One ; 6(12): e28797, 2011.
Article in English | MEDLINE | ID: mdl-22194915

ABSTRACT

Angiogenin (ANG) acts on both vascular endothelial cells and cancer cells, but the underlying mechanism remains elusive. In this study, we carried out a co-immunoprecipitation assay in HeLa cells and identified 14 potential ANG-interacting proteins. Among these proteins, ß-actin, α-actinin 4, and non-muscle myosin heavy chain 9 are stress fiber components and involved in cytoskeleton organization and movement, which prompted us to investigate the mechanism of action of ANG in cell migration. Upon confirmation of the interactions between ANG and the three proteins, further studies revealed that ANG co-localized with ß-actin and α-actinin 4 at the leading edge of migrating cells. Down-regulation of ANG resulted in fewer but thicker stress fibers with less dynamics, which was associated with the enlargements of focal adhesions. The focal adhesion kinase activity and cell migration capacity were significantly decreased in ANG-deficient cells. Taken together, our data demonstrated that the existence of ANG in the cytoplasm optimizes stress fiber assembly and focal adhesion formation to accommodate cell migration. The finding that ANG promoted cancer cell migration might provide new clues for tumor metastasis research.


Subject(s)
Cell Movement , Focal Adhesions/metabolism , Ribonuclease, Pancreatic/metabolism , Stress Fibers/metabolism , Cytoskeleton/metabolism , Down-Regulation , Enzyme Activation , Focal Adhesion Protein-Tyrosine Kinases/metabolism , HeLa Cells , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mass Spectrometry , Molecular Sequence Annotation , Phosphorylation , Protein Binding , Pseudopodia/metabolism , Ribonuclease, Pancreatic/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL