Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(4): 113972, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38517892

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1) is a transcriptional activator that mediates cellular adaptation to decreased oxygen availability. HIF-1 recruits chromatin-modifying enzymes leading to changes in histone acetylation, citrullination, and methylation at target genes. Here, we demonstrate that hypoxia-inducible gene expression in estrogen receptor (ER)-positive MCF7 and ER-negative SUM159 human breast cancer cells requires the histone H2A/H2B chaperone facilitates chromatin transcription (FACT) and the H2B ubiquitin ligase RING finger protein 20/40 (RNF20/40). Knockdown of FACT or RNF20/40 expression leads to decreased transcription initiation and elongation at HIF-1 target genes. Mechanistically, FACT and RNF20/40 are recruited to hypoxia response elements (HREs) by HIF-1 and stabilize binding of HIF-1 (and each other) at HREs. Hypoxia induces the monoubiquitination of histone H2B at lysine 120 at HIF-1 target genes in an HIF-1-dependent manner. Together, these findings delineate a cooperative molecular mechanism by which FACT and RNF20/40 stabilize multiprotein complex formation at HREs and mediate histone ubiquitination to facilitate HIF-1 transcriptional activity.


Subject(s)
DNA-Binding Proteins , Hypoxia-Inducible Factor 1 , Ubiquitin-Protein Ligases , Humans , Cell Hypoxia , Cell Line, Tumor , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Histones/metabolism , Hypoxia-Inducible Factor 1/metabolism , MCF-7 Cells , Protein Binding , Response Elements , Transcription Factors/metabolism , Transcriptional Activation , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
2.
Cell Rep ; 42(3): 112164, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36857181

ABSTRACT

Intratumoral hypoxia is a microenvironmental feature that promotes breast cancer progression and is associated with cancer mortality. Plexin B3 (PLXNB3) is highly expressed in estrogen receptor-negative breast cancer, but the underlying mechanisms and consequences have not been thoroughly investigated. Here, we report that PLXNB3 expression is increased in response to hypoxia and that PLXNB3 is a direct target gene of hypoxia-inducible factor 1 (HIF-1) in human breast cancer cells. PLXNB3 expression is correlated with HIF-1α immunohistochemistry, breast cancer grade and stage, and patient mortality. Mechanistically, PLXNB3 is required for hypoxia-induced MET/SRC/focal adhesion kinase (FAK) and MET/SRC/STAT3/NANOG signaling as well as hypoxia-induced breast cancer cell migration, invasion, and cancer stem cell specification. PLXNB3 knockdown impairs tumor formation and lung metastasis in orthotopic breast cancer mouse models.


Subject(s)
Breast Neoplasms , Lung Neoplasms , Animals , Female , Humans , Mice , Breast Neoplasms/pathology , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung Neoplasms/pathology , Neoplastic Stem Cells/metabolism
3.
Sci Adv ; 8(49): eabo5000, 2022 12 09.
Article in English | MEDLINE | ID: mdl-36490339

ABSTRACT

Hypoxia is a key characteristic of the breast cancer microenvironment that promotes expression of the transcriptional activator hypoxia-inducible factor 1 (HIF-1) and is associated with poor patient outcome. HIF-1 increases the expression or activity of stem cell pluripotency factors, which control breast cancer stem cell (BCSC) specification and are required for cancer metastasis. Here, we identify nuclear prelamin A recognition factor (NARF) as a hypoxia-inducible, HIF-1 target gene in human breast cancer cells. NARF functions as an essential coactivator by recruiting the histone demethylase KDM6A to OCT4 bound to genes encoding the pluripotency factors NANOG, KLF4, and SOX2, leading to demethylation of histone H3 trimethylated at lysine-27 (H3K27me3), thereby increasing the expression of NANOG, KLF4, and SOX2, which, together with OCT4, mediate BCSC specification. Knockdown of NARF significantly decreased the BCSC population in vitro and markedly impaired tumor initiation capacity and lung metastasis in orthotopic mouse models.


Subject(s)
Breast Neoplasms , Hypoxia-Inducible Factor 1 , Animals , Female , Humans , Mice , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Histones/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Microenvironment/genetics , Tumor Microenvironment/physiology , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism
4.
Cancer Lett ; 531: 1-13, 2022 04 10.
Article in English | MEDLINE | ID: mdl-35101541

ABSTRACT

Lung cancer is the most common cancer and the leading cause of cancer deaths worldwide. In addition to coding genes, the contribution of long noncoding RNA (lncRNA) to non-small cell lung cancer (NSCLC) remains unclear. Here, we explored lncRNA expression profiles by Affymetrix Gene Chip Human Transcriptome Array 2.0 in 37 paired samples of tumorous NSCLC tissues and adjacent nontumorous tissues. We showed that LHFPL3-AS2 is a novel lncRNA, significantly decreased in NSCLC tissues. LHFPL3-AS2 was further validated in an additional 93 paired samples of NSCLC. Low levels of LHFPL3-AS2 expression were highly correlated with poor overall survival, TNM stage, and metastasis of NSCLC patients. Enhanced expression of LHFPL3-AS2 inhibited NSCLC invasion and metastasis in vitro and in vivo. Moreover, downregulation of LHFPL3-AS2 reduced its specific interaction with SFPQ, resulting in more SFPQ binding to the promoter of TXNIP and causing the transcriptional repression of TXNIP, thus finally promoting the migration and invasion of NSCLC cells. Furthermore, LHFPL3-AS2 was shown to be regulated by EGR1 under hypoxia.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , RNA, Long Noncoding , Carcinoma, Non-Small-Cell Lung/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , Membrane Proteins , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
5.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Article in English | MEDLINE | ID: mdl-33952697

ABSTRACT

Breast cancer patients with increased expression of hypoxia-inducible factors (HIFs) in primary tumor biopsies are at increased risk of metastasis, which is the major cause of breast cancer-related mortality. The mechanisms by which intratumoral hypoxia and HIFs regulate metastasis are not fully elucidated. In this paper, we report that exposure of human breast cancer cells to hypoxia activates epidermal growth factor receptor (EGFR) signaling that is mediated by the HIF-dependent expression of a disintegrin and metalloprotease 12 (ADAM12), which mediates increased ectodomain shedding of heparin-binding EGF-like growth factor, an EGFR ligand, leading to EGFR-dependent phosphorylation of focal adhesion kinase. Inhibition of ADAM12 expression or activity decreased hypoxia-induced breast cancer cell migration and invasion in vitro, and dramatically impaired lung metastasis after orthotopic implantation of MDA-MB-231 human breast cancer cells into the mammary fat pad of immunodeficient mice.


Subject(s)
ADAM12 Protein/genetics , ADAM12 Protein/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Hypoxia/metabolism , ADAM12 Protein/deficiency , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , ErbB Receptors/metabolism , Female , Gene Knockdown Techniques , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Lung/pathology , Lung Neoplasms/pathology , Mice , Mice, SCID , Neoplasm Metastasis/genetics , Signal Transduction , Tumor Microenvironment
6.
Nat Commun ; 12(1): 295, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436560

ABSTRACT

Circular RNAs (circRNA) are a class of covalently closed single-stranded RNAs that have been implicated in cancer progression. Here we identify circNDUFB2 to be downregulated in non-small cell lung cancer (NSCLC) tissues, and to negatively correlate with NSCLC malignant features. Elevated circNDUFB2 inhibits growth and metastasis of NSCLC cells. Mechanistically, circNDUFB2 functions as a scaffold to enhance the interaction between TRIM25 and IGF2BPs, a positive regulator of tumor progression and metastasis. This TRIM25/circNDUFB2/IGF2BPs ternary complex facilitates ubiquitination and degradation of IGF2BPs, with this effect enhanced by N6-methyladenosine (m6A) modification of circNDUFB2. Moreover, circNDUFB2 is also recognized by RIG-I to activate RIG-I-MAVS signaling cascades and recruit immune cells into the tumor microenvironment (TME). Our data thus provide evidences that circNDUFB2 participates in the degradation of IGF2BPs and activation of anti-tumor immunity during NSCLC progression via the modulation of both protein ubiquitination and degradation, as well as cellular immune responses.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Disease Progression , Lung Neoplasms/genetics , Lung Neoplasms/immunology , RNA, Circular/metabolism , RNA-Binding Proteins/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , DEAD Box Protein 58/metabolism , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Neoplasm Metastasis , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Stability , Proteolysis , RNA, Circular/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Immunologic , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
7.
Hepatology ; 73(2): 644-660, 2021 02.
Article in English | MEDLINE | ID: mdl-32298475

ABSTRACT

BACKGROUND AND AIMS: Peroxisome proliferator-activated receptor-gamma (PPARγ) coactivator-1α (PGC1α) is a key regulator of mitochondrial biogenesis and respiration. PGC1α is involved in the carcinogenesis, progression, and metabolic state of cancer. However, its role in the progression of hepatocellular carcinoma (HCC) remains unclear. APPROACH AND RESULTS: In this study, we observed that PGC1α was down-regulated in human HCC. A clinical study showed that low levels of PGC1α expression were correlated with poor survival, vascular invasion, and larger tumor size. PGC1α inhibited the migration and invasion of HCC cells with both in vitro experiments and in vivo mouse models. Mechanistically, PGC1α suppressed the Warburg effect through down-regulation of pyruvate dehydrogenase kinase isozyme 1 (PDK1) mediated by the WNT/ß-catenin pathway, and inhibition of the WNT/ß-catenin pathway was induced by activation of PPARγ. CONCLUSIONS: Low levels of PGC1α expression indicate a poor prognosis for HCC patients. PGC1α suppresses HCC metastasis by inhibiting aerobic glycolysis through regulating the WNT/ß-catenin/PDK1 axis, which depends on PPARγ. PGC1α is a potential factor for predicting prognosis and a therapeutic target for HCC patients.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/secondary , Liver Neoplasms/pathology , Lung Neoplasms/secondary , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Biomarkers, Tumor/blood , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Liver/pathology , Liver/surgery , Liver Neoplasms/blood , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/blood , Prognosis , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Warburg Effect, Oncologic , Wnt Signaling Pathway/genetics , Xenograft Model Antitumor Assays
8.
Nat Commun ; 10(1): 3200, 2019 07 19.
Article in English | MEDLINE | ID: mdl-31324812

ABSTRACT

Circular RNAs (circRNAs) are identified as vital regulators in a variety of cancers. However, the role of circRNA in lung squamous cell carcinoma (LUSC) remains largely unknown. Herein, we explore the expression profiles of circRNA and mRNA in 5 paired samples of LUSC. By analyzing the co-expression network of differentially expressed circRNAs and dysregulated mRNAs, we identify that a cell cycle-related circRNA, circTP63, is upregulated in LUSC tissues and its upregulation is correlated with larger tumor size and higher TNM stage in LUSC patients. Elevated circTP63 promotes cell proliferation both in vitro and in vivo. Mechanistically, circTP63 shares miRNA response elements with FOXM1. circTP63 competitively binds to miR-873-3p and prevents miR-873-3p to decrease the level of FOXM1, which upregulates CENPA and CENPB, and finally facilitates cell cycle progression.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Disease Progression , Forkhead Box Protein M1/metabolism , Gene Expression Regulation, Neoplastic , RNA, Circular/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation , Animals , Carcinoma, Squamous Cell/genetics , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation , Centromere Protein A/metabolism , Centromere Protein B/metabolism , Female , Gene Expression Profiling , Gene Regulatory Networks , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice, Inbred BALB C , MicroRNAs , Middle Aged , Neoplasms, Experimental , RNA, Circular/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcriptome , Tumor Suppressor Proteins/genetics
9.
Acta Biochim Biophys Sin (Shanghai) ; 51(3): 263-276, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30883650

ABSTRACT

Metastasis is the main reason for high recurrence and poor survival of hepatocellular carcinoma (HCC). The molecular mechanism underlying HCC metastasis remains unclear. In this study, we found that argininosuccinate synthase 1 (ASS1) expression was significantly decreased and down-regulation of ASS1 was closely correlated with poor prognosis in HCC patients. DNA methylation led to the down-regulation of ASS1 in HCC. Stable silencing of ASS1 promoted migration and invasion of HCC cells, whereas overexpression of ASS1-inhibited metastasis of HCC cells in vivo and in vitro. We also revealed that ASS1-knockdown increased the phosphorylation level of S727STAT3, which contributed to HCC metastasis by up-regulation of inhibitor of differentiation 1 (ID1). These findings indicate that ASS1 inhibits HCC metastasis and may serve as a target for HCC diagnosis and treatment.


Subject(s)
Argininosuccinate Synthase/physiology , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Argininosuccinate Synthase/antagonists & inhibitors , Argininosuccinate Synthase/genetics , Carcinoma, Hepatocellular/enzymology , Cell Line, Tumor , Cell Movement , DNA Methylation , Humans , Inhibitor of Differentiation Protein 1/genetics , Liver Neoplasms/enzymology , Mice , Neoplasm Invasiveness , Neoplasm Metastasis/prevention & control , STAT3 Transcription Factor/physiology , Signal Transduction/physiology
10.
J Cancer ; 10(4): 918-926, 2019.
Article in English | MEDLINE | ID: mdl-30854098

ABSTRACT

Alterations in cellular metabolism are one of the characteristics in cancer. They are not only the result of tumor progression but also the cause of cancer initiation. Pyruvate dehydrogenase kinase 4 (PDK4) is a key metabolic enzyme, which regulates cell metabolism by inhibiting pyruvate dehydrogenase (PDH). However, the function and regulating mechanism of PDK4 in HCC remain unclear. Here, we found that the expression of PDK4 was significantly decreased in HCC tissues, and its downregulation could predict poor prognosis of HCC patients. Silencing PDK4 significantly facilitated proliferation and migration of HCC cells. Knockdown of PDK4 didn't influence the oxidative phosphorylation and glycolysis capacity of HCC cells in vitro. However, knockdown of PDK4 increased expression of key lipogenic enzymes, fatty acid synthase (FASN) and stearoyl-CoA desaturase (SCD), which finally induced lipogenesis. These data suggest that PDK4 inhibits proliferation and migration of HCC cells probably via suppressing lipogenesis.

11.
Cancer Lett ; 450: 98-109, 2019 05 28.
Article in English | MEDLINE | ID: mdl-30790682

ABSTRACT

Long noncoding RNAs (lncRNAs) are implicated as novel drivers in hepatocellular carcinoma (HCC), but the underlying mechanisms of this relationship with hepatocarcinogenesis are unknown. We report a novel, liver-specific lncRNA LINC01093 that shows significant downregulation in HCC tissues. LINC01093 expression is inversely correlated with cancer embolus and HCC TNM stage and as a prognostic predictor for HCC patients. LINC01093 overexpression significantly suppresses HCC cell proliferation and metastasis in vitro and in vivo. Conversely, its knockdown promotes HCC progression. Mechanistic analyses indicate that LINC01093 directly binds insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1), interfering with interaction between IGF2BP1 and glioma-associated oncogene homolog 1 (GLI1) mRNA. The result is degradation of GLI1 mRNA, further affecting expression of GLI1 downstream molecules involved in HCC progression. The liver-enriched lncRNA LINC01093 is a promising prognostic indicator for HCC patients, and the newly identified LINC01093-IGF2BP1-GLI1 axis shows potential for therapeutic targets in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Zinc Finger Protein GLI1/genetics , Animals , Carcinogenesis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Down-Regulation , Genes, Tumor Suppressor , Heterografts , Humans , Liver Neoplasms/metabolism , Mice , Neoplasm Metastasis , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Zinc Finger Protein GLI1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...