Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Am Soc Mass Spectrom ; 34(8): 1692-1700, 2023 Aug 02.
Article in English | MEDLINE | ID: mdl-37463068

ABSTRACT

Arginine methylation catalyzed by protein arginine methyltransferases (PRMTs) is a prevalent post-translational modification (PTM) that regulates diverse cellular processes. Aberrant expression of type I PRMTs that catalyze asymmetric arginine dimethylation (ADMA) is often found in cancer, though little is known about the ADMA status of substrate proteins in tumors. Using LC-MS/MS along with pan-specific ADMA antibodies, we performed global mapping of ADMA in five patient-derived xenograft (PDX) tumors representing different subtypes of human breast cancer. In total, 403 methylated sites from 213 proteins were identified, including 322 novel sites when compared to the PhosphositesPlus database. Moreover, using peptide arrays in vitro, approximately 70% of the putative substrates were validated to be methylated by PRMT1, PRMT4, and PRMT6. Notably, when compared with our previously identified ADMA sites from breast cancer cell lines, only 75 ADMA sites overlapped between cell lines and PDX tumors. Collectively, this study provides a useful resource for both PRMT and breast cancer communities for further exploitation of the functions of PRMT dysregulation during breast cancer progression.


Subject(s)
Breast Neoplasms , Proteome , Humans , Female , Chromatography, Liquid , Arginine/metabolism , Tandem Mass Spectrometry , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Nuclear Proteins
3.
Redox Biol ; 56: 102452, 2022 10.
Article in English | MEDLINE | ID: mdl-36084349

ABSTRACT

PURPOSE: Bile acids are steroid synthesized in liver, which are essential for fat emulsification, cholesterol excretion and gut microbial homeostasis. However, the role of bile acids in leukemia progression remains unclear. We aim at exploring the effects and mechanisms of chenodeoxycholic acid (CDCA), a type of bile acids, on acute myeloid leukemia (AML) progression. RESULTS: Here, we found that CDCA was decreased in feces and plasma of AML patients, positively correlated with the diversity of gut microbiota, and negatively associated with AML prognosis. We further demonstrated that CDCA suppressed AML progression both in vivo and in vitro. Mechanistically, CDCA bound to mitochondria to cause mitochondrial morphology damage containing swelling and reduction of cristae, decreased mitochondrial membrane potential and elevated mitochondrial calcium level, which resulted in the production of excessive reactive oxygen species (ROS). Elevated ROS further activated p38 MAPK signaling pathway, which collaboratively promoted the accumulation of lipid droplets (LDs) through upregulating the expression of the diacylglycerol O-acyltransferase 1 (DGAT1). As the consequence of the abundance of ROS and LDs, lipid peroxidation was enhanced in AML cells. Moreover, we uncovered that CDCA inhibited M2 macrophage polarization and suppressed the proliferation-promoting effects of M2 macrophages on AML cells in co-cultured experiments. CONCLUSION: Our findings demonstrate that CDCA suppresses AML progression through synergistically promoting LDs accumulation and lipid peroxidation via ROS/p38 MAPK/DGAT1 pathway caused by mitochondrial dysfunction in leukemia cells and inhibiting M2 macrophage polarization.


Subject(s)
Leukemia, Myeloid, Acute , p38 Mitogen-Activated Protein Kinases , Bile Acids and Salts/metabolism , Calcium/metabolism , Chenodeoxycholic Acid/metabolism , Chenodeoxycholic Acid/pharmacology , Cholesterol/metabolism , Diacylglycerol O-Acyltransferase/metabolism , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Lipid Peroxidation , Macrophages/metabolism , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Blood ; 139(7): 1066-1079, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34699595

ABSTRACT

Mutations in chromatin regulator ASXL1 are frequently identified in myeloid malignancies, in particular ∼40% of patients with chronic myelomonocytic leukemia (CMML). ASXL1 mutations are associated with poor prognosis in CMML and significantly co-occur with NRAS mutations. Here, we show that concurrent ASXL1 and NRAS mutations defined a population of CMML patients who had shorter leukemia-free survival than those with ASXL1 mutation only. Corroborating this human data, Asxl1-/- accelerated CMML progression and promoted CMML transformation to acute myeloid leukemia (AML) in NrasG12D/+ mice. NrasG12D/+;Asxl1-/- (NA) leukemia cells displayed hyperactivation of MEK/ERK signaling, increased global levels of H3K27ac, upregulation of Flt3. Moreover, we find that NA-AML cells overexpressed all the major inhibitory immune checkpoint ligands: programmed death-ligand 1 (PD-L1)/PD-L2, CD155, and CD80/CD86. Among them, overexpression of PD-L1 and CD86 correlated with upregulation of AP-1 transcription factors (TFs) in NA-AML cells. An AP-1 inhibitor or short hairpin RNAs against AP-1 TF Jun decreased PD-L1 and CD86 expression in NA-AML cells. Once NA-AML cells were transplanted into syngeneic recipients, NA-derived T cells were not detectable. Host-derived wild-type T cells overexpressed programmed cell death protein 1 (PD-1) and T-cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT) receptors, leading to a predominant exhausted T-cell phenotype. Combined inhibition of MEK and BET resulted in downregulation of Flt3 and AP-1 expression, partial restoration of the immune microenvironment, enhancement of CD8 T-cell cytotoxicity, and prolonged survival in NA-AML mice. Our study suggests that combined targeted therapy and immunotherapy may be beneficial for treating secondary AML with concurrent ASXL1 and NRAS mutations.


Subject(s)
Disease Models, Animal , GTP Phosphohydrolases/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Myelomonocytic, Chronic/pathology , Membrane Proteins/genetics , Mutation , Repressor Proteins/genetics , Tumor Microenvironment , Animals , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/immunology , Mice , Monomeric GTP-Binding Proteins/genetics , Phenotype , Signal Transduction
6.
EMBO J ; 40(22): e108065, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34487377

ABSTRACT

The pyruvate kinase M2 isoform (PKM2) is preferentially expressed in cancer cells to regulate anabolic metabolism. Although PKM2 was recently reported to regulate lipid homeostasis, the molecular mechanism remains unclear. Herein, we discovered an ER transmembrane protein 33 (TMEM33) as a downstream effector of PKM2 that regulates activation of SREBPs and lipid metabolism. Loss of PKM2 leads to up-regulation of TMEM33, which recruits RNF5, an E3 ligase, to promote SREBP-cleavage activating protein (SCAP) degradation. TMEM33 is transcriptionally regulated by nuclear factor erythroid 2-like 1 (NRF1), whose cleavage and activation are controlled by PKM2 levels. Total plasma cholesterol levels are elevated by either treatment with PKM2 tetramer-promoting agent TEPP-46 or by global PKM2 knockout in mice, highlighting the essential function of PKM2 in lipid metabolism. Although depletion of PKM2 decreases cancer cell growth, global PKM2 knockout accelerates allografted tumor growth. Together, our findings reveal the cell-autonomous and systemic effects of PKM2 in lipid homeostasis and carcinogenesis, as well as TMEM33 as a bona fide regulator of lipid metabolism.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lipid Metabolism/physiology , Membrane Proteins/metabolism , Thyroid Hormones/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cholesterol/blood , Female , Gene Expression Regulation, Neoplastic , Homeostasis , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice, Knockout , Sterol Regulatory Element Binding Protein 1/metabolism , Thyroid Hormones/genetics , Xenograft Model Antitumor Assays , Thyroid Hormone-Binding Proteins
7.
Mol Cell ; 81(6): 1276-1291.e9, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33539787

ABSTRACT

Aberrant cell proliferation is a hallmark of cancer, including glioblastoma (GBM). Here we report that protein arginine methyltransferase (PRMT) 6 activity is required for the proliferation, stem-like properties, and tumorigenicity of glioblastoma stem cells (GSCs), a subpopulation in GBM critical for malignancy. We identified a casein kinase 2 (CK2)-PRMT6-regulator of chromatin condensation 1 (RCC1) signaling axis whose activity is an important contributor to the stem-like properties and tumor biology of GSCs. CK2 phosphorylates and stabilizes PRMT6 through deubiquitylation, which promotes PRMT6 methylation of RCC1, which in turn is required for RCC1 association with chromatin and activation of RAN. Disruption of this pathway results in defects in mitosis. EPZ020411, a specific small-molecule inhibitor for PRMT6, suppresses RCC1 arginine methylation and improves the cytotoxic activity of radiotherapy against GSC brain tumor xenografts. This study identifies a CK2α-PRMT6-RCC1 signaling axis that can be therapeutically targeted in the treatment of GBM.


Subject(s)
Brain Neoplasms , Carcinogenesis , Cell Cycle Proteins , Glioblastoma , Guanine Nucleotide Exchange Factors , Mitosis/radiation effects , Neoplasm Proteins , Nuclear Proteins , Protein-Arginine N-Methyltransferases , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/radiotherapy , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/radiation effects , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/radiotherapy , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Male , Mice , Mitosis/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Signal Transduction/genetics , Signal Transduction/radiation effects , Xenograft Model Antitumor Assays
8.
J Proteome Res ; 17(8): 2744-2754, 2018 08 03.
Article in English | MEDLINE | ID: mdl-29923408

ABSTRACT

As a popular sample preparation approach, filter-aided sample preparation (FASP) has been widely used in proteomic analysis. However, several limitations have been noted, including sample loss during filtration, repetitive centrifugation steps, and the possibility of breakage of filtration membrane. Extraction bias among different sample preparation strategies presents another challenge. To overcome these limitations and address remaining challenges, we developed a novel surfactant and chaotropic agent assisted sequential extraction/on-pellet digestion (SCAD) protocol. The new strategy resulted in higher protein yield and improved peptide recovery and protein coverage compared to two conventional sample preparation methods (FASP and urea). In combination of three strategies, more than 10,000 distinct protein groups were identified with 1% FDR from MDA-MB-231 cells without any prefractionation. This in-depth proteome analysis was accomplished by optimization of protein extraction, enzymatic digestion, LC gradient, and peptide sequencing method. Ingenuity Pathways Analysis (IPA) of proteins exclusively identified in SCAD revealed several crucial signaling pathways that regulate breast cancer progression. SCAD also enabled an unbiased extraction of different categories of proteins (membrane, intracellular, nuclear) associated with tumorigenesis, which integrates the advantages of FASP and urea extraction. This novel strategy expedites comprehensive protein identification, which is applicable for biomarker discovery in various types of cancers.


Subject(s)
Proteins/metabolism , Proteomics/methods , Specimen Handling/methods , Surface-Active Agents/chemistry , Amino Acid Sequence , Breast Neoplasms/metabolism , Carcinogenesis , Cell Line, Tumor , Female , Humans , Proteins/analysis , Proteins/isolation & purification , Proteolysis , Signal Transduction , Urea
9.
Analyst ; 143(11): 2508-2519, 2018 May 29.
Article in English | MEDLINE | ID: mdl-29687791

ABSTRACT

The system-wide site-specific analysis of intact glycopeptides is crucial for understanding the exact functional relevance of protein glycosylation. A dedicated workflow with the capability to simultaneously characterize and quantify intact glycopeptides in a site-specific and high-throughput manner is essential to reveal specific glycosylation alteration patterns in complex biological systems. In this study, an enhanced, dedicated, large-scale site-specific quantitative N-glycoproteomics workflow has been established, which includes improved specific extraction of membrane-bound glycoproteins using the filter aided sample preparation (FASP) method, enhanced enrichment of N-glycopeptides using sequential hydrophilic interaction liquid chromatography (HILIC) and multi-lectin affinity (MLA) enrichment, site-specific N-glycopeptide characterization enabled by EThcD, relative quantitation utilizing isobaric N,N-dimethyl leucine (DiLeu) tags and automated FDR-based large-scale data analysis by Byonic. For the first time, our study shows that HILIC complements to a very large extent to MLA enrichment with only 20% overlapping in enriching intact N-glycopeptides. When applying the developed workflow to site-specific N-glycoproteome study in PANC1 cells, we were able to identify 1067 intact N-glycopeptides, representing 311 glycosylation sites and 88 glycan compositions from 205 glycoproteins. We further applied this approach to study the glycosylation alterations in PKM2 knockout cells vs. parental breast cancer cells and revealed altered N-glycoprotein/N-glycopeptide patterns and very different glycosylation microheterogeneity for different types of glycans. To obtain a more comprehensive map of glycoprotein alterations, N-glycopeptides after treatment with PNGase F were also analyzed. A total of 484 deglycosylated peptides were quantified, among which 81 deglycosylated peptides from 70 glycoproteins showed significant changes. KEGG pathway analysis revealed that the PI3K/Akt signaling pathway was highly enriched, which provided evidence to support the previous finding that PKM2 knockdown cancer cells rely on activation of Akt for their survival. With glycosylation being one of the most important signaling modulators, our results provide additional evidence that signaling pathways are closely regulated by metabolism.


Subject(s)
Breast Neoplasms/chemistry , Glycopeptides/analysis , Glycoproteins/analysis , Carrier Proteins/genetics , Cell Line, Tumor , Electrons , Gene Knockout Techniques , Glycosylation , Humans , Leucine , Membrane Proteins/genetics , Thyroid Hormones/genetics , Thyroid Hormone-Binding Proteins
10.
Mol Pharmacol ; 93(3): 197-207, 2018 03.
Article in English | MEDLINE | ID: mdl-29295894

ABSTRACT

The biologic effects of estrogens are transduced by two estrogen receptors (ERs), ERα and ERß, which function in dimer forms. The ERα/α homodimer promotes and the ERß/ß inhibits estrogen-dependent growth of mammary epithelial cells; the functions of ERα/ß heterodimers remain elusive. Using compounds that promote ERα/ß heterodimerization, we have previously shown that ERα/ß heterodimers appeared to inhibit tumor cell growth and migration in vitro. Further dissection of ERα/ß heterodimer functions was hampered by the lack of ERα/ß heterodimer-specific ligands. Herein, we report a multistep workflow to identify the selective ERα/ß heterodimer-inducing compound. Phytoestrogenic compounds were first screened for ER transcriptional activity using reporter assays and ER dimerization preference using a bioluminescence resonance energy transfer assay. The top hits were subjected to in silico modeling to identify the pharmacophore that confers ERα/ß heterodimer specificity. The pharmacophore encompassing seven features that are potentially important for the formation of the ERα/ß heterodimer was retrieved and subsequently used for virtual screening of large chemical libraries. Four chemical compounds were identified that selectively induce ERα/ß heterodimers over their respective homodimers. Such ligands will become unique tools to reveal the functional insights of ERα/ß heterodimers.


Subject(s)
Computational Biology/methods , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Mammary Glands, Human/cytology , Phytoestrogens/pharmacology , Bioluminescence Resonance Energy Transfer Techniques , Cell Line , Drug Evaluation, Preclinical , Estrogen Receptor alpha/chemistry , Estrogen Receptor beta/chemistry , Female , HEK293 Cells , Humans , Ligands , MCF-7 Cells , Mammary Glands, Human/metabolism , Models, Molecular , Phytoestrogens/chemistry , Protein Multimerization
12.
Nat Cell Biol ; 19(11): 1358-1370, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29058718

ABSTRACT

Metabolic reprogramming is a hallmark of cancer. Herein we discover that the key glycolytic enzyme pyruvate kinase M2 isoform (PKM2), but not the related isoform PKM1, is methylated by co-activator-associated arginine methyltransferase 1 (CARM1). PKM2 methylation reversibly shifts the balance of metabolism from oxidative phosphorylation to aerobic glycolysis in breast cancer cells. Oxidative phosphorylation depends on mitochondrial calcium concentration, which becomes critical for cancer cell survival when PKM2 methylation is blocked. By interacting with and suppressing the expression of inositol-1,4,5-trisphosphate receptors (InsP3Rs), methylated PKM2 inhibits the influx of calcium from the endoplasmic reticulum to mitochondria. Inhibiting PKM2 methylation with a competitive peptide delivered by nanoparticles perturbs the metabolic energy balance in cancer cells, leading to a decrease in cell proliferation, migration and metastasis. Collectively, the CARM1-PKM2 axis serves as a metabolic reprogramming mechanism in tumorigenesis, and inhibiting PKM2 methylation generates metabolic vulnerability to InsP3R-dependent mitochondrial functions.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carrier Proteins/metabolism , Glycolysis/physiology , Guanylate Cyclase/metabolism , Membrane Proteins/metabolism , Thyroid Hormones/metabolism , Animals , Calcium/metabolism , Cell Line , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Endoplasmic Reticulum/metabolism , Female , HEK293 Cells , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , MCF-7 Cells , Methylation , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/metabolism , Neoplasm Metastasis/pathology , Oxidative Phosphorylation , Thyroid Hormone-Binding Proteins
13.
Nat Commun ; 8: 15571, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28537268

ABSTRACT

Protein arginine methyltransferases (PRMTs) introduce arginine methylation, a post-translational modification with the increasingly eminent role in normal physiology and disease. PRMT4 or coactivator-associated arginine methyltransferase 1 (CARM1) is a propitious target for cancer therapy; however, few CARM1 substrates are known, and its mechanism of substrate recognition is poorly understood. Here we employed a quantitative mass spectrometry approach to globally profile CARM1 substrates in breast cancer cell lines. We identified >130 CARM1 protein substrates and validated in vitro >90% of sites they encompass. Bioinformatics analyses reveal enrichment of proline-containing motifs, in which both methylation sites and their proximal sequences are frequently targeted by somatic mutations in cancer. Finally, we demonstrate that the N-terminus of CARM1 is involved in substrate recognition and nearly indispensable for substrate methylation. We propose that development of CARM1-specific inhibitors should focus on its N-terminus and predict that other PRMTs may employ similar mechanism for substrate recognition.


Subject(s)
Amino Acid Motifs/physiology , Protein Processing, Post-Translational/physiology , Protein-Arginine N-Methyltransferases/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Arginine/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Chromatography, High Pressure Liquid/methods , Computational Biology , Enzyme Inhibitors , Female , Gene Knockout Techniques , HEK293 Cells , Humans , MCF-7 Cells , Methylation , Molecular Targeted Therapy/methods , Mutation , Proline/chemistry , Proline/metabolism , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/chemistry , Protein-Arginine N-Methyltransferases/genetics , Substrate Specificity/physiology , Tandem Mass Spectrometry/methods
14.
Biochem Biophys Res Commun ; 484(3): 565-571, 2017 03 11.
Article in English | MEDLINE | ID: mdl-28132807

ABSTRACT

The oncoprotein hepatitis B X-interacting protein (HBXIP) results in the dysregulation of lipid metabolism to enhance the development of breast cancer. Acyl-CoA synthetase long-chain family member 1 (ACSL1) is required for thioesterification of long-chain fatty acids into their acyl-CoA derivatives. In this study, we present a hypothesis that HBXIP might be involved in the regulation of ACSL1 in breast cancer. Interestingly, we found that the overexpression of HBXIP was able to up-regulate ACSL1 at the levels of mRNA and protein in a dose-dependent manner in breast cancer cells. Conversely, silencing of HBXIP led to the opposite results. Mechanistically, HBXIP as a coactivator interacted with transcriptional factor Sp1 through binding to the promoter of ACSL1 by ChIP assays analysis, leading to the transcription of ACSL1 in breast cancer cells. Immunohistochemistry staining revealed that the positive rate of ACSL1 was 71.4% (35/49) in clinical breast cancer tissues, HBXIP 79.6% (39/49), in which the positive rate of ACSL1 was 76.9% (30/39) in the HBXIP-positive specimens. But, few positive rate of ACSL1 10% (1/10) was observed in normal breast tissues. The mRNA levels of ACSL1 were significantly higher in clinical breast cancer tissues than those in their corresponding peritumor tissues. The mRNA levels of ACSL1 were positively associated with those of HBXIP in clinical breast cancer tissues. Thus, we conclude that the oncoprotein HBXIP is able to up-regulate ACSL1 through activating the transcriptional factor Sp1 in breast cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Coenzyme A Ligases/metabolism , Gene Expression Regulation, Neoplastic , Sp1 Transcription Factor/metabolism , Cell Line, Tumor , Humans , Signal Transduction , Transcriptional Activation , Up-Regulation
15.
Cancer Res ; 76(16): 4696-707, 2016 08 15.
Article in English | MEDLINE | ID: mdl-26980761

ABSTRACT

Abnormal lipid metabolism is a hallmark of tumorigenesis. Accumulating evidence demonstrates that fatty acid synthase (FAS, FASN) is a metabolic oncogene that supports the growth and survival of tumor cells and is highly expressed in many cancers. Here, we report that the oncoprotein, hepatitis B X-interacting protein (HBXIP, LAMTOR5) contributes to abnormal lipid metabolism. We show that high expression of HBXIP in 236 breast cancer patients was significantly associated with decreased overall survival and progression-free survival. Interestingly, the expression of HBXIP was positively related to that of FAS in clinical breast cancer tissues, and HBXIP overexpression in breast cancer cells resulted in FAS upregulation. Mechanistically, HBXIP upregulated SREBP-1c (SREBF1), which activates the transcription of FAS, by directly interacting with and coactivating nuclear receptor (NR) liver X receptors (LXR). Physiologically, LXRs are activated via a coactivator containing NR motif in a ligand-dependent manner. However, in breast cancer cells, HBXIP containing the corepressor/nuclear receptor motif with special flanking sequence could coactivate LXRs independent of ligand. Moreover, overexpressed SREBP-1c was able to activate the transcription of HBXIP, forming a positive-feedback loop. Functionally, HBXIP enhanced lipogenesis, resulting in the growth of breast cancer cells in vitro and in vivo Thus, we conclude that the oncoprotein HBXIP contributes to the abnormal lipid metabolism in breast cancer through LXRs/SREBP-1c/FAS signaling, providing new insights into the mechanisms by which cancer cells reprogram lipid metabolism in their favor. Cancer Res; 76(16); 4696-707. ©2016 AACR.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/pathology , Lipid Metabolism/physiology , Liver X Receptors/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , fas Receptor/metabolism , Animals , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Proliferation/physiology , Disease-Free Survival , Electrophoretic Mobility Shift Assay , Female , Gene Expression Regulation, Neoplastic/physiology , Heterografts , Humans , Immunoprecipitation , Kaplan-Meier Estimate , Mice, Inbred BALB C , Microscopy, Confocal , Polymerase Chain Reaction , Signal Transduction/physiology , Tissue Array Analysis
16.
Oncotarget ; 7(1): 241-54, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26540633

ABSTRACT

Highly up-regulated in liver cancer (HULC) is a long non-coding RNA (lncRNA). We found that HULC up-regulated sphingosine kinase 1 (SPHK1), which is involved in tumor angiogenesis. Levels of HULC were positively correlated with levels of SPHK1 and its product, sphingosine-1-phosphate (S1P), in patients HCC samples. HULC increased SPHK1 in hepatoma cells. Chicken chorioallantoic membrane (CAM) assays revealed that si-SPHK1 remarkably blocked the HULC-enhanced angiogenesis. Mechanistically, HULC activated the promoter of SPHK1 in hepatoma cells through the transcription factor E2F1. Chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift assay (EMSA) further showed that E2F1 was capable of binding to the E2F1 element in the SPHK1 promoter. HULC increased the expression of E2F1 in hepatoma cells and levels of HULC were positively correlated with those of E2F1 in HCC tissues. Intriguingly, HULC sequestered miR-107, which targeted E2F1 mRNA 3'UTR, by complementary base pairing. Functionally, si-SPHK1 remarkably abolished the HULC-enhanced tumor angiogenesis in vitro and in vivo. Taken together, we conclude that HULC promotes tumor angiogenesis in liver cancer through miR-107/E2F1/SPHK1 signaling. Our finding provides new insights into the mechanism of tumor angiogenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Neovascularization, Pathologic/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA, Long Noncoding/genetics , 3' Untranslated Regions/genetics , Adult , Aged , Animals , Blotting, Western , Chick Embryo , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Female , Hep G2 Cells , Humans , Liver Neoplasms/blood supply , Male , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , Neovascularization, Physiologic/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Up-Regulation/genetics
17.
Oncotarget ; 6(29): 27199-213, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26309161

ABSTRACT

The glucose metabolism reprogramming is a hallmark of cancer. The oncoprotein hepatitis B X-interacting protein (HBXIP) functions in the development of breast cancer. In this study, we supposed that HBXIP might be involved in the glucose metabolism reprogramming in breast cancer. We showed that HBXIP led to increases in generation of intracellular glucose and lactate, as well as decreases in generation of reactive oxygen species. Expression of synthesis of cytochrome c oxidase 2 (SCO2) and pyruvate dehydrogenase alpha 1 (PDHA1), two factors of metabolic switch from oxidative phosphorylation to aerobic glycolysis, was suppressed by HBXIP. In addition, miR-183/182 and miR-96 directly inhibited the expression of SCO2 and PDHA1 through targeting their mRNA coding sequences (CDSs), respectively. Interestingly, HBXIP elevated the miR-183/96/182 cluster expression through hypoxia-inducible factor 1α (HIF1α). The stability of HIF1α was enhanced by HBXIP through disassociating interaction of von Hippel-Lindau protein (pVHL) with HIF1α. Moreover, miR-183 increased the levels of HIF1α protein through directly targeting CDS of VHL mRNA, forming a feedback loop of HIF1α/miR-183/pVHL/HIF1α. In function, HBXIP-elevated miR-183/96/182 cluster enhanced the glucose metabolism reprogramming in vitro. HBXIP-triggered glucose metabolism reprogramming promoted the growth of breast cancer in vivo. Thus, we conclude that the oncoprotein HBXIP enhances glucose metabolism reprogramming through suppressing SCO2 and PDHA1 in breast cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Glucose/metabolism , Mitochondrial Proteins/metabolism , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Molecular Chaperones , Neoplasm Transplantation , Oxygen/chemistry , Phosphorylation , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction
18.
Acta Pharmacol Sin ; 35(9): 1207-14, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25087998

ABSTRACT

AIM: MiR-506 is a miRNA involved in carcinogenesis of several kinds of cancer. In this study, we explored whether miR-506 played a critical role in hepatocellular carcinoma (HCC). METHODS: Twenty HCC and adjacent normal liver tissue samples were collected. Human hepatoma cell lines HepG2 and H7402 were used for in vitro studies. The expression of miR-506 and transcriptional co-activator YAP was examined using qRT-PCR. Western blot analysis was used to measure the expression of YAP and its target genes. Luciferase reporter gene assay was used to identify YAP as a target gene of miR-506. MTT and EdU assays were carried out for functional analysis. RESULTS: The expression of miR-506 was significantly lower in HCC than in adjacent normal liver tissues. Bioinformatics analysis revealed that YAP mRNA might be one of the targets of miR-506, and miR-506 in HCC tissues was found to be negatively correlated with YAP (r=-0.605). In both HepG2 and H7402 cells, miR-506 dose-dependently down-regulated YAP and its target genes c-Myc and the connective tissue growth factor (CTGF). Luciferase reporter gene assays demonstrated that miR-506 targeted the wild type 3'UTR of YAP mRNA, but not a 3'UTR with a mutant seed site. Furthermore, miR-506 significantly inhibited the proliferation of HepG2 and H7402 cells, while anti-miR-506 enhanced the cell proliferation, which was blocked by YAP siRNA. CONCLUSION: MiR-506 suppresses the proliferation of hepatoma cells by targeting YAP mRNA.


Subject(s)
3' Untranslated Regions/genetics , Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Hepatocellular/genetics , Cell Proliferation/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Phosphoproteins/genetics , RNA, Messenger/genetics , Cell Line, Tumor , Hep G2 Cells , Humans , Transcription Factors , YAP-Signaling Proteins
19.
Mol Cancer ; 13: 128, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24886421

ABSTRACT

BACKGROUND: Hepatitis B virus X protein (HBx) plays crucial roles in hepatocarcinogenesis. However, the underlying mechanism remains elusive. We have reported that HBx is able to up-regulate survivin in hepatocellular carcinoma tissues. The oncopreotein hepatitis B X-interacting protein (HBXIP), a target of miR-520b, is involved in the development of cancer. In this study, we focus on the investigation of hepatocarcinogenesis mediated by HBx. METHODS: The expression of HBx and survivin was examined in the liver tissues of HBx-Tg mice. The effect of HBx/survivin on the growth of LO2-X-S cells was determined by colony formation and transplantation in nude mice. The effect of HBx/survivin on promoter of miR-520b was determined by Western blot analysis, luciferase reporter gene assay, co-immunoprecipitation (co-IP) and chromatin immunoprecipitation (ChIP), respectively. The expression of HBx, survivin and HBXIP was detected by immunohistochemistry and real-time PCR in clinical HCC tissues, respectively. The DNA demethylation of HBXIP promoter was examined. The functional influence of miR-520b and HBXIP on proliferation of hepatoma cells was analyzed by MTT, colony formation, EdU and transplantation in nude mice in vitro and in vivo. RESULTS: In this study, we provided evidence that HBx up-regulated survivin in the liver cancer tissues of HBx-Tg mice aged 18 M. The engineered LO2 cell lines with survivin and/or HBx were successfully established, termed LO2-X-S. MiR-520b was down-regulated in LO2-X-S cells and clinical HCC tissues. Our data revealed that HBx survivin-dependently down-regulated miR-520b through interacting with Sp1 in the cells. HBXIP was highly expressed in LO2-X-S cells, liver cancer tissues of HBx-Tg mice aged 18 M and clinical HCC tissues (75.17%, 112/149). The expression level of HBXIP was positively associated with those of HBx or survivin in clinical HCC tissues. In addition, we showed that HBx survivin-dependently up-regulated HBXIP through inducing demethylation of HBXIP promoter in LO2-X-S cells and clinical HCC tissues. In function, low level miR-520b and high level HBXIP mediated by HBx with partner survivin contributed to the growth of LO2-X-S cells in vitro and in vivo. CONCLUSION: HBx accelerates hepatocarcinogenesis with partner survivin through modulating tumor suppressor miR-520b and oncoprotein HBXIP.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Hepatocellular/genetics , Hepacivirus/genetics , Inhibitor of Apoptosis Proteins/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Trans-Activators/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , DNA Methylation , Gene Expression Regulation , Hepacivirus/metabolism , Hepatitis B/complications , Hepatitis B/genetics , Hepatitis B/metabolism , Hepatitis B/pathology , Host-Pathogen Interactions , Humans , Inhibitor of Apoptosis Proteins/metabolism , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Nude , Mice, Transgenic , MicroRNAs/metabolism , Neoplasm Transplantation , Promoter Regions, Genetic , Signal Transduction , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Survivin , Trans-Activators/metabolism , Viral Regulatory and Accessory Proteins
20.
Cancer Lett ; 352(2): 169-78, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24882622

ABSTRACT

Hepatitis B X-interacting protein (HBXIP) is an important oncoprotein in hepatocarcinogenesis. Here, we found that the expression levels of HBXIP were positively associated with those of Secretogranin III (SCG3) in clinical hepatocellular carcinoma tissues. We identified that HBXIP up-regulated the expression of SCG3 through modulating both E2F transcription factor 1 (E2F1) and miR-509-3p. HBXIP suppressed miR-509-3p through activating NF-κB. In function, we showed that SCG3 increased the proliferation of hepatoma cells and HBXIP enhanced the proliferation of the cells via SCG3 in vitro and in vivo. Thus, we conclude that HBXIP facilitates the proliferation of hepatoma cells through up-regulating SCG3.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Hepatocellular/metabolism , Chromogranins/metabolism , E2F1 Transcription Factor/metabolism , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding Sites , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Chromogranins/genetics , E2F1 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , NF-kappa B/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/metabolism , Signal Transduction , Time Factors , Transfection , Tumor Burden , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...