Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Mol Cell ; 71(2): 306-318.e7, 2018 07 19.
Article in English | MEDLINE | ID: mdl-30017583

ABSTRACT

DNA N6-methyladenine (6mA) modification is the most prevalent DNA modification in prokaryotes, but whether it exists in human cells and whether it plays a role in human diseases remain enigmatic. Here, we showed that 6mA is extensively present in the human genome, and we cataloged 881,240 6mA sites accounting for ∼0.051% of the total adenines. [G/C]AGG[C/T] was the most significantly associated motif with 6mA modification. 6mA sites were enriched in the coding regions and mark actively transcribed genes in human cells. DNA 6mA and N6-demethyladenine modification in the human genome were mediated by methyltransferase N6AMT1 and demethylase ALKBH1, respectively. The abundance of 6mA was significantly lower in cancers, accompanied by decreased N6AMT1 and increased ALKBH1 levels, and downregulation of 6mA modification levels promoted tumorigenesis. Collectively, our results demonstrate that DNA 6mA modification is extensively present in human cells and the decrease of genomic DNA 6mA promotes human tumorigenesis.


Subject(s)
Adenine/analogs & derivatives , AlkB Homolog 1, Histone H2a Dioxygenase/metabolism , Genome, Human , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism , Adenine/metabolism , AlkB Homolog 1, Histone H2a Dioxygenase/genetics , Animals , Carcinogenesis/genetics , DNA/genetics , DNA Methylation , Heterografts , Humans , Mice , Mice, Nude , Site-Specific DNA-Methyltransferase (Adenine-Specific)/genetics
2.
Mol Cell ; 68(1): 171-184.e6, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28985503

ABSTRACT

A substantial fraction of eukaryotic transcripts are considered long non-coding RNAs (lncRNAs), which regulate various hallmarks of cancer. Here, we discovered that the lncRNA HOXB-AS3 encodes a conserved 53-aa peptide. The HOXB-AS3 peptide, not lncRNA, suppresses colon cancer (CRC) growth. Mechanistically, the HOXB-AS3 peptide competitively binds to the ariginine residues in RGG motif of hnRNP A1 and antagonizes the hnRNP A1-mediated regulation of pyruvate kinase M (PKM) splicing by blocking the binding of the ariginine residues in RGG motif of hnRNP A1 to the sequences flanking PKM exon 9, ensuring the formation of lower PKM2 and suppressing glucose metabolism reprogramming. CRC patients with low levels of HOXB-AS3 peptide have poorer prognoses. Our study indicates that the loss of HOXB-AS3 peptide is a critical oncogenic event in CRC metabolic reprogramming. Our findings uncover a complex regulatory mechanism of cancer metabolism reprogramming orchestrated by a peptide encoded by an lncRNA.


Subject(s)
Cell Transformation, Neoplastic/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Peptides/genetics , RNA, Long Noncoding/genetics , Alternative Splicing , Amino Acid Motifs , Animals , Binding, Competitive , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Exons , HeLa Cells , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Peptides/antagonists & inhibitors , Peptides/metabolism , Protein Binding , Protein Interaction Mapping , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
3.
Mol Ther ; 27(10): 1718-1725, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31526596

ABSTRACT

Non-coding RNAs (ncRNAs) are unique RNA transcripts that have been widely identified in the eukaryotic genome and have been shown to play key roles in the development of many cancers. However, the rapid development of genome-wide translation profiling and ribosome profiling has revealed that a small number of small open reading frames (sORFs) within ncRNAs actually have peptide- or protein-coding potential. The peptides or proteins encoded by ncRNA (HOXB-AS3, encoded by long ncRNA [lncRNA]; FBXW7-185aa, PINT-87aa, and SHPRH-146aa, encoded by circular RNA [circRNA]; and miPEP-200a and miPEP-200b, encoded by primary miRNAs) have been shown to be critical players in cancer development and progression, through effects upon the regulation of glucose metabolism, the epithelial-to-mesenchymal transition, and the ubiquitination pathway. In this review, we summarize the reported peptides or proteins encoded by ncRNAs in cancer and explore the application of these peptides or proteins in the development of anti-tumor drugs and the identification of relevant therapeutic targets and tumor biomarkers.


Subject(s)
Gene Regulatory Networks , Neoplasms/genetics , RNA, Untranslated/metabolism , Disease Progression , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Humans , Neoplasms/metabolism , Peptides/genetics , Proteins/genetics , Ubiquitination
4.
AAPS PharmSciTech ; 21(1): 6, 2019 Nov 21.
Article in English | MEDLINE | ID: mdl-31754916

ABSTRACT

The aim of the study is to investigate the feasibility of fabricating FDM 3D-printed gastric floating tablets with low infill percentages and the effect of infill percentage on the properties of gastric floating tablets in vitro. Propranolol hydrochloride was selected as a model drug, and drug-loaded polyvinyl alcohol (PVA) filaments were produced by hot melt extrusion (HME). Ellipsoid-shaped gastric floating tablets with low infill percentage of 15% and 25% (namely E-15 and E-25) were then prepared respectively by feeding the extruded filaments to FDM 3D printer. Thermogravimetric analysis (TGA), differential scanning calorimetry (DSC), X-ray powder diffraction (XRD), and scanning electron microscopy (SEM) were employed to characterize the filaments and 3D-printed tablets, and a series of evaluations were performed to the 3D-printed tablets, including the weight variation, drug content, hardness, in vitro floating behavior, and drug release of the tablets. The SEM results showed that the drug-loaded filaments and 3D-printed tablets appeared intact without defects, and the printed tablets were composed of filaments deposited uniformly layer by layer. The model drug and the excipients were thermally stable under the process temperature of extruding and printing, with a small amount of drug crystals dispersing in the drug-loaded filaments and 3D-printed tablets. Both E-15 and E-25 could float on artificial gastric fluids without any lag time and released in a sustained manner. Compared with E-15, the E-25 presented less weight variation, higher tablet hardness, shorter floating time, and longer drug release time.


Subject(s)
Drug Carriers/chemical synthesis , Excipients/chemical synthesis , Printing, Three-Dimensional , Tablets/chemical synthesis , Technology, Pharmaceutical/methods , Calorimetry, Differential Scanning/methods , Drug Carriers/pharmacokinetics , Drug Liberation , Excipients/pharmacokinetics , Polyvinyl Alcohol/chemical synthesis , Polyvinyl Alcohol/pharmacokinetics , Propranolol/chemical synthesis , Propranolol/pharmacokinetics , Tablets/pharmacokinetics , X-Ray Diffraction/methods
5.
J Pathol ; 239(2): 186-96, 2016 06.
Article in English | MEDLINE | ID: mdl-26969828

ABSTRACT

The tricho-rhino-phalangeal syndrome 1 gene (TRPS1), which was initially found to be associated with tricho-rhino-phalangeal syndrome, is critical for the development and differentiation of bone, hair follicles and kidney. However, its role in cancer progression is largely unknown. In this study, we demonstrated that down-regulation of TRPS1 correlated with distant metastasis, tumour recurrence and poor survival rate in cancer patients. TRPS1 was frequently down-regulated in high-metastatic cancer cell lines from the breast, colon and nasopharynx. Silencing of TRPS1 stimulated epithelial-mesenchymal transition (EMT), migration and invasion in vitro and metastasis in vivo, while TRPS1 over-expression exhibited the opposite effects. Using quantitative proteomics, FOXA1, a negative regulator of epithelial-mesenchymal transition (EMT), was shown to be down-regulated by TRPS1 knockdown. Ectopic expression of FOXA1 blocked the enhancement of EMT, migration and invasion induced by TRPS1 silencing. Mechanistically, TRPS1, acting as a transcription activator, directly induced FOXA1 transcription by binding to the FOXA1 promoter. We further showed that down-regulation of TRPS1 was induced by miR-373 binding to the 3' UTR of TRPS1. Over-expression of TRPS1, but not TRPS1 3' UTR, blocked the enhancement of migration and invasion induced by miR-373. Taken together, we consider that down-regulation of TRPS1 by miR-373, acting as a transcriptional activator, promotes EMT and metastasis by repressing FOXA1 transcription, expanding upon its previously reported role as a transcription repressor. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Breast Neoplasms/genetics , Colonic Neoplasms/genetics , DNA-Binding Proteins/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , MicroRNAs/genetics , Nasopharyngeal Neoplasms/genetics , Transcription Factors/metabolism , 3' Untranslated Regions/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Differentiation , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA-Binding Proteins/genetics , Down-Regulation , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Repressor Proteins , Transcription Factors/genetics
6.
Bioinformatics ; 31(4): 608-9, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25677125

ABSTRACT

SUMMARY: Disease ontology (DO) annotates human genes in the context of disease. DO is important annotation in translating molecular findings from high-throughput data to clinical relevance. DOSE is an R package providing semantic similarity computations among DO terms and genes which allows biologists to explore the similarities of diseases and of gene functions in disease perspective. Enrichment analyses including hypergeometric model and gene set enrichment analysis are also implemented to support discovering disease associations of high-throughput biological data. This allows biologists to verify disease relevance in a biological experiment and identify unexpected disease associations. Comparison among gene clusters is also supported. AVAILABILITY AND IMPLEMENTATION: DOSE is released under Artistic-2.0 License. The source code and documents are freely available through Bioconductor (http://www.bioconductor.org/packages/release/bioc/html/DOSE.html). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online. CONTACT: gcyu@connect.hku.hk or tqyhe@jnu.edu.cn.


Subject(s)
Computational Biology/methods , Disease/genetics , Gene Ontology , Programming Languages , Semantics , Software , Databases, Genetic , Humans , Multigene Family
7.
J Pathol ; 236(2): 175-85, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25678401

ABSTRACT

Amplification of the activated Cdc42-associated kinase 1 (ACK1) gene is frequent in gastric cancer (GC). However, little is known about the clinical roles and molecular mechanisms of ACK1 abnormalities in GC. Here, we found that the ACK1 protein level and ACK1 phosphorylation at Tyr 284 were frequently elevated in GC and associated with poor patient survival. Ectopic ACK1 expression in GC cells induced epithelial-mesenchymal transition (EMT) and promoted migration and invasion in vitro, and metastasis in vivo; the depletion of ACK1 induced the opposite effects. We utilized SILAC quantitative proteomics to discover that the level of the cell cycle-related protein ecdysoneless homologue (ECD) was markedly altered by ACK1. Overexpression of ECD promoted EMT, migration, and invasion in GC, similar to the effects of ACK1 overexpression. Silencing of ECD completely blocked the augmentation of ACK1 overexpression-induced EMT, migration, and invasion. Mechanistically, ACK1 phosphorylated AKT at Thr 308 and Ser 473 and activated the AKT pathway to up-regulate the transcription factor POU2F1, which directly bound to the promoter region of its novel target gene ECD and thus regulated ECD expression in GC cells. Furthermore, the phosphorylation levels of AKT at Thr 308 and Ser 473 and POU2F1 and ECD levels were positively associated with ACK1 levels in clinical GC specimens. Collectively, we have demonstrated that ACK1 promotes EMT, migration, and invasion by activating AKT-POU2F1-ECD signalling in GC cells. ACK1 may be employed as a new prognostic factor and therapeutic target for GC.


Subject(s)
Carrier Proteins/metabolism , Epithelial-Mesenchymal Transition/physiology , Octamer Transcription Factor-1/metabolism , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/physiopathology , Adult , Aged , Animals , Cell Communication/physiology , Cell Line, Tumor , Female , Gene Knockdown Techniques , Heterografts , Humans , Male , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Prognosis , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Stomach Neoplasms/metabolism , Up-Regulation
8.
Front Med ; 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38907157

ABSTRACT

RNA modification is an essential component of the epitranscriptome, regulating RNA metabolism and cellular functions. Several types of RNA modifications have been identified to date; they include N6-methyladenosine (m6A), N1-methyladenosine (m1A), 5-methylcytosine (m5C), N7-methylguanosine (m7G), N6,2'-O-dimethyladenosine (m6Am), N4-acetylcytidine (ac4C), etc. RNA modifications, mediated by regulators including writers, erasers, and readers, are associated with carcinogenesis, tumor microenvironment, metabolic reprogramming, immunosuppression, immunotherapy, chemotherapy, etc. A novel perspective indicates that regulatory subunits and post-translational modifications (PTMs) are involved in the regulation of writer, eraser, and reader functions in mediating RNA modifications, tumorigenesis, and anticancer therapy. In this review, we summarize the advances made in the knowledge of different RNA modifications (especially m6A) and focus on RNA modification regulators with functions modulated by a series of factors in cancer, including regulatory subunits (proteins, noncoding RNA or peptides encoded by long noncoding RNA) and PTMs (acetylation, SUMOylation, lactylation, phosphorylation, etc.). We also delineate the relationship between RNA modification regulator functions and carcinogenesis or cancer progression. Additionally, inhibitors that target RNA modification regulators for anticancer therapy and their synergistic effect combined with immunotherapy or chemotherapy are discussed.

9.
Proteomics ; 13(21): 3222-32, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23970500

ABSTRACT

Isolated from Elephantopus scaber L., a Chinese medicinal herb that is widely used to prevent and treat cancers in China, isodeoxyelephantopin (ESI) exerted antitumor effects on several cancer cells. However, its antitumor mechanism is still not clear. In this study, we found that ESI could induce G2/M arrest and subsequently stimulate cell apoptosis in dose- and time-dependent manners. We used SILAC quantitative proteomics to identify ESI-regulated proteins in cancer cells, and found that 124 proteins were significantly altered in expression. Gene ontology and Ingenuity Pathway Analysis revealed that these proteins were mainly involved in the regulation of oxidative stress and inflammation response. Functional studies demonstrated that ESI induced G2/M arrest and apoptosis by inducing ROS generation, and that antioxidant N-acetyl-l-cysteine could block the ESI-induced antitumor effects. Accumulated ROS resulted in DNA breakage, subsequent G2/M arrest and mitochondrial-mediated apoptosis. ESI upregulated the expression of anticancer inflammation factors IL-12a, IFN-α, and IFN-ß through ROS-dependent and independent pathways. The current work reveals that ESI exerts its antitumor effects through ROS-dependent DNA damage, mitochondrial-mediated apoptosis mechanism and antitumor inflammation factor pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Lactones/pharmacology , Nasopharyngeal Neoplasms/metabolism , Proteome/drug effects , Proteomics/methods , Sesquiterpenes/pharmacology , Apoptosis/drug effects , Carcinoma , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cytokines/analysis , Cytokines/metabolism , Humans , Nasopharyngeal Carcinoma , Protein Interaction Maps , Proteome/analysis , Proteome/chemistry
10.
Proteomics ; 13(1): 169-78, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23161516

ABSTRACT

Andrographolide-lipoic acid conjugate (AL-1) is a new in-house synthesized chemical entity, which was derived by covalently linking andrographolide with lipoic acid. However, its anti-cancer effect and cytotoxic mechanism remains unknown. In this study, we found that AL-1 could significantly inhibit cell viability of human leukemia K562 cells by inducing G2/M arrest and apoptosis in a dose-dependent manner. Thirty-one AL-1-regulated protein alterations were identified by proteomics analysis. Gene ontology and ingenuity pathway analysis revealed that a cluster of proteins of oxidative redox state and apoptotic cell death-related proteins, such as PRDX2, PRDX3, PRDX6, TXNRD1, and GLRX3, were regulated by AL-1. Functional studies confirmed that AL-1 induced apoptosis of K562 cells through a ROS-dependent mechanism, and anti-oxidant, N-acetyl-L-cysteine, could completely block AL-1-induced cytotoxicity, implicating that ROS generation played a vital role in AL-1 cytotoxicity. Accumulated ROS resulted in oxidative DNA damage and subsequent G2/M arrest and mitochondrial-mediated apoptosis. The current work reveals that a novel andrographolide derivative AL-1 exerts its anticancer cytotoxicity through a ROS-dependent DNA damage and mitochondrial-mediated apoptosis mechanism.


Subject(s)
Apoptosis/drug effects , Diterpenes/pharmacology , Proteome/analysis , Thioctic Acid/pharmacology , Cell Survival/drug effects , DNA Damage/drug effects , Diterpenes/chemistry , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , K562 Cells , Leukemia/drug therapy , Leukemia/metabolism , Leukemia/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Thioctic Acid/chemistry
11.
Nat Commun ; 14(1): 3815, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37369679

ABSTRACT

N6-methyladenosine (m6A) modification plays important roles in bioprocesses and diseases. AlkB homolog 5 (ALKBH5) is one of two m6A demethylases. Here, we reveal that ALKBH5 is acetylated at lysine 235 (K235) by lysine acetyltransferase 8 and deacetylated by histone deacetylase 7. K235 acetylation strengthens the m6A demethylation activity of ALKBH5 by increasing its recognition of m6A on mRNA. RNA-binding protein paraspeckle component 1 (PSCP1) is a regulatory subunit of ALKBH5 and preferentially interacts with K235-acetylated ALKBH5 to recruit and facilitate the recognition of m6A mRNA by ALKBH5, thereby promoting m6A erasure. Mitogenic signals promote ALKBH5 K235 acetylation. K235 acetylation of ALKBH5 is upregulated in cancers and promotes tumorigenesis. Thus, our findings reveal that the m6A demethylation activity of ALKBH5 is orchestrated by its K235 acetylation and regulatory subunit PSPC1 and that K235 acetylation is necessary for the m6A demethylase activity and oncogenic roles of ALKBH5.


Subject(s)
Carcinogenesis , Cell Transformation, Neoplastic , Humans , Acetylation , RNA, Messenger/metabolism , Carcinogenesis/genetics , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , Demethylation , RNA-Binding Proteins/metabolism
12.
Proteomics ; 12(14): 2391-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22887948

ABSTRACT

Genistein exerts its anticarcinogenic effects by inducing G2/M arrest and apoptosis of cancer cells. However, the precise molecular mechanism of action of genistein has not been completely elucidated. In this study, we used quantitative proteomics to identify the genistein-induced protein alterations in gastric cancer cells and investigate the molecular mechanism responsible for the anti-cancer actions of genistein. Total 86 proteins were identified to be regulated by genistein, most of which were clustered into the regulation of cell division and G2/M transition, consistent with the anti-cancer effect of genistein. Many proteins including kinesin family proteins, TPX2, CDCA8, and CIT were identified for the first time to be regulated by genistein. Interestingly, five kinesin family proteins including KIF11, KIF20A, KIF22, KIF23, and CENPF were found to be simultaneously downregulated by genistein. Significantly decreased KIF20A was selected for further functional studies. The silencing of KIF20A inhibited cell viability and induced G2/M arrest, similar to the effects of genistein treatment in gastric cancer. And the silencing of KIF20A also increased cancer cell sensitivity to genistein inhibition, whereas overexpression of KIF20A markedly attenuated genistein-induced cell viability inhibition and G2/M arrest. These observations suggested that KIF20A played an important role in anti-cancer actions of genistein, and thus may be a potential molecular target for drug intervention of gastric cancer.


Subject(s)
Genistein/pharmacology , Kinesins/metabolism , Mitosis/drug effects , Proteome/drug effects , Stomach Neoplasms/drug therapy , Anticarcinogenic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Gene Regulatory Networks/drug effects , Humans , Kinesins/genetics , Protein Interaction Maps/drug effects , Proteome/analysis , RNA Interference , RNA, Small Interfering , Signal Transduction/drug effects , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
13.
Proteomics ; 11(5): 912-20, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21271679

ABSTRACT

Although microRNAs (miRNAs) have been reported to play an important role in carcinogenesis, their molecular mechanism remains largely unknown because of our limited understanding of miRNA target genes. miR-373 was found to be capable of promoting breast cancer invasion and metastasis, but only a target gene was experimentally identified on the basis of mRNA expression analysis. In this study, we used SILAC-based quantitative proteomics to globally identify the genes regulated by miR-373. Totally, 3666 proteins were identified, and 335 proteins were found to be regulated by miR-373. Among the 192 proteins that were downregulated by miR-373, 27 (14.1%) were predicted to have at least one potential match site at their 3'-UTR for miR-373 seed sequence. However, miR-373 did not affect the mRNA level of the five selected candidate targets, TXNIP, TRPS1, RABEP1, GRHL2 and HIP1, suggesting that the protein expressions were regulated by miR-373 via translational inhibition instead of mRNA degradation. Luciferase and mutation assays validated that TXNIP and RABEP1 were the direct target genes of miR-373. More than 30 proteins reported to be involved in cancer invasion and metastasis were found to be regulated by miR-373 in breast cancer for the first time.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation , MicroRNAs/metabolism , Proteomics/methods , Vesicular Transport Proteins/metabolism , 3' Untranslated Regions , Base Sequence , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cell Movement , Databases, Genetic , Down-Regulation , Female , Genes, Reporter , Genome-Wide Association Study , Humans , Luciferases/genetics , Luciferases/metabolism , MicroRNAs/genetics , Molecular Sequence Data , Mutation , Neoplasm Metastasis , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vesicular Transport Proteins/genetics
14.
Proteomics ; 11(18): 3657-64, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21751384

ABSTRACT

We previously used proteomics technology to globally identify gastric cancer-associated proteins and found that gastrokine 1 (GKN1) was dramatically underexpressed in gastric cancer tissues. Here, we further showed that GKN1 could inhibit cell growth and induce cell cycle arrest in gastric cancer cells. The activity of protein kinase PKCδ/θ was inhibited by GKN1, whereas the activity of ERK1/2 and JNK1/2 was increased by GKN1, suggesting that GKN1 induced growth inhibition of gastric cancer cells by synergistically regulating the activity of these protein kinases. Seventy-four proteins were found to be regulated by GKN1 by proteomics analysis, including α-enolase (ENO1) and Cathepsin D. Interestingly, ENO1 is an important hub in the protein-protein interaction network of the 74 differential proteins. Silencing of ENO1 resulted in growth inhibition and cell cycle arrest of gastric cancer cells, similar to the effect of GKN1 overexpression in cells, whereas ENO1 overexpression blocked GKN1-induced growth inhibition and cell cycle arrest. These observations suggested that ENO1 downregulation played an important role in GKN1-induced growth inhibition of gastric cancer cells.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Neoplastic , Peptide Hormones/metabolism , Proteomics/methods , Stomach Neoplasms/metabolism , Apoptosis , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Cycle Checkpoints , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Databases, Protein , Down-Regulation , Electrophoresis, Gel, Two-Dimensional , Enzyme Activation , Flow Cytometry , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Peptide Hormones/genetics , Phosphopyruvate Hydratase/metabolism , Phosphorylation , Protein Interaction Maps , RNA, Small Interfering/metabolism , Transfection , Tumor Suppressor Proteins/metabolism
15.
Proteomics ; 10(5): 976-86, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20049867

ABSTRACT

Genistein is a natural protein tyrosine kinase inhibitor that exerts anti-cancer effect by inducing G2/M arrest and apoptosis. However, the phosphotyrosine signaling pathways mediated by genistein are largely unknown. In this study, we combined tyrosine phosphoprotein enrichment with MS-based quantitative proteomics technology to globally identify genistein-regulated tyrosine phosphoproteins aiming to depict genistein-inhibited phosphotyrosine cascades. Our experiments resulted in the identification of 213 phosphotyrosine sites on 181 genistein-regulated proteins. Many identified phosphoproteins, including nine protein kinases, eight receptors, five protein phosphatases, seven transcriptical regulators and four signal adaptors, were novel inhibitory effectors with no previously known function in the anti-cancer mechanism of genistein. Functional analysis suggested that genistein-regulated protein tyrosine phosphorylation mainly by inhibiting the activity of tyrosine kinase EGFR, PDGFR, insulin receptor, Abl, Fgr, Itk, Fyn and Src. Core signaling molecules inhibited by genistein can be functionally categorized into the canonial Receptor-MAPK or Receptor-PI3K/AKT cascades. The method used here may be suitable for the identification of inhibitory effectors and tyrosine kinases regulated by anti-cancer drugs.


Subject(s)
Genistein/pharmacology , Phosphoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , Proteomics/methods , Signal Transduction/drug effects , Amino Acid Sequence , Cell Line, Tumor , Down-Regulation/drug effects , G2 Phase/drug effects , Humans , Isotope Labeling , Mass Spectrometry , Mitosis/drug effects , Molecular Sequence Data , Peptides/chemistry , Phosphoproteins/chemistry , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/analysis , Reproducibility of Results
16.
Theranostics ; 10(19): 8558-8572, 2020.
Article in English | MEDLINE | ID: mdl-32754263

ABSTRACT

Rationale: Hepatocellular carcinoma (HCC) is one of the most lethal cancers, and few molecularly targeted anticancer therapies have been developed to treat it. Thus, the identification of new therapeutic targets is urgent. Metabolic reprogramming is an important hallmark of cancer. However, how ubiquitin ligases are involved in the regulation of cancer metabolism remains poorly understood. Methods: RT-PCR, western blot and IHC were used to determine ZFP91 expression. RNAi, cell proliferation, colony formation and transwell assays were used to determine the in vitro functions of ZFP91. Mouse xenograft models were used to study the in vivo effects of ZFP91. Co-IP together with mass spectrometry or western blot was utilized to investigate protein-protein interaction. Ubiquitination was analyzed using IP together with western blot. RNA splicing was assessed by using RT-PCR followed by restriction digestion. Lactate production and glucose uptake assays were used to analyze cancer metabolism. Results: We identified that an E3 ligase zinc finger protein 91 (ZFP91) suppressed HCC metabolic reprogramming, cell proliferation and metastasis in vitro and in vivo. Mechanistically, ZFP91 promoted the Lys48-linked ubiquitination of the oncoprotein hnRNP A1 at lysine 8 and proteasomal degradation, thereby inhibiting hnRNP A1-dependent PKM splicing, subsequently resulting in higher PKM1 isoform formation and lower PKM2 isoform formation and suppressing HCC glucose metabolism reprogramming, cell proliferation and metastasis. Moreover, HCC patients with lower levels of ZFP91 have poorer prognoses, and ZFP91 is an independent prognostic factor for patients with HCC. Conclusions: Our study identifies ZFP91 as a tumor suppressor of hepatocarcinogenesis and HCC metabolism reprogramming and proposes it as a novel prognostic biomarker and therapeutic target of HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Carrier Proteins/genetics , Liver Neoplasms/pathology , Membrane Proteins/genetics , RNA Splicing , Thyroid Hormones/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Neoplasm Staging , Neoplasm Transplantation , Prognosis , Signal Transduction , Survival Analysis , Ubiquitination , Thyroid Hormone-Binding Proteins
17.
Adv Sci (Weinh) ; 7(10): 1903233, 2020 May.
Article in English | MEDLINE | ID: mdl-32440474

ABSTRACT

Conventional therapies for late-stage colorectal cancer (CRC) have limited effects because of chemoresistance, recurrence, and metastasis. The "hidden" proteins/peptides encoded by long noncoding RNAs (lncRNAs) may be a novel resource bank for therapeutic options for patients with cancer. Here, lncRNA LOC90024 is discovered to encode a small 130-amino acid protein that interacts with several splicing regulators, such as serine- and arginine-rich splicing factor 3 (SRSF3), to regulate mRNA splicing, and the protein thus is named "Splicing Regulatory Small Protein" (SRSP). SRSP, but not LOC90024 lncRNA itself, promotes CRC tumorigenesis and progression, while silencing of SRSP suppresses CRC tumorigenesis. Mechanistically, SRSP increases the binding of SRSF3 to exon 3 of transcription factor Sp4, resulting in the inclusion of Sp4 exon 3 to induce the formation of the "cancerous" long Sp4 isoform (L-Sp4 protein) and inhibit the formation of the "noncancerous" short Sp4 isoform (S-Sp4 peptide), which lacks the transactivation domain. The upregulated SRSP level is positively associated with malignant phenotypes and poor prognosis in patients with CRC. Collectively, the findings uncover that a lncRNA-encoded small protein SRSP induces "cancerous" Sp4 splicing variant formation and may be a potential prognostic biomarker and therapeutic target for patients with CRC.

18.
Nat Commun ; 11(1): 1685, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32245947

ABSTRACT

N6-methyladenosine (m6A) is the most prevalent modification in eukaryotic RNAs. The biological importance of m6A relies on m6A readers, which control mRNA fate and function. However, it remains unexplored whether additional regulatory subunits of m6A readers are involved in the m6A recognition on RNAs. Here we discover that the long noncoding RNA (lncRNA) LINC00266-1 encodes a 71-amino acid peptide. The peptide mainly interacts with the RNA-binding proteins, including the m6A reader IGF2BP1, and is thus named "RNA-binding regulatory peptide" (RBRP). RBRP binds to IGF2BP1 and strengthens m6A recognition by IGF2BP1 on RNAs, such as c-Myc mRNA, to increase the mRNA stability and expression of c-Myc, thereby promoting tumorigenesis. Cancer patients with RBRPhigh have a poor prognosis. Thus, the oncopeptide RBRP encoded by LINC00266-1 is a regulatory subunit of m6A readers and strengthens m6A recognition on the target RNAs by the m6A reader to exert its oncogenic functions.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , RNA Processing, Post-Transcriptional/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Animals , Binding Sites , Cell Line, Tumor , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Methylation , Mice , Middle Aged , Prognosis , Proto-Oncogene Proteins c-myc/genetics , RNA Stability/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/metabolism , Survival Analysis , Tissue Array Analysis , Xenograft Model Antitumor Assays
19.
Theranostics ; 9(3): 676-690, 2019.
Article in English | MEDLINE | ID: mdl-30809301

ABSTRACT

Cancer cells undergo metabolic reprogramming to support their energy demand and biomass synthesis. However, the mechanisms driving cancer metabolism reprogramming are not well understood. Methods: The differential proteins and interacted proteins were identified by proteomics. Western blot, qRT-PCR and IHC staining were used to analyze TBC1D8 levels. In vivo tumorigenesis and metastasis were performed by xenograft tumor model. Cross-Linking assays were designed to analyze PKM2 polymerization. Lactate production, glucose uptake and PK activity were determined. Results: We established two aggressive ovarian cancer (OVCA) cell models with increased aerobic glycolysis. TBC1D8, a member of the TBC domain protein family, was significantly up-regulated in the more aggressive OVCA cells. TBC1D8 is amplified and up-regulated in OVCA tissues. OVCA patients with high TBC1D8 levels have poorer prognoses. TBC1D8 promotes OVCA tumorigenesis and aerobic glycolysis in a GAP activity-independent manner in vitro and in vivo. TBC1D8 bound to PKM2, not PKM1, via its Rab-GAP TBC domain. Mechanistically, TBC1D8 binds to PKM2 and hinders PKM2 tetramerization to decreases pyruvate kinase activity and promote aerobic glycolysis, and to promote the nuclear translocation of PKM2, which induces the expression of genes which are involved in glucose metabolism and cell cycle. Conclusions:TBC1D8 drives OVCA tumorigenesis and metabolic reprogramming, and TBC1D8 serves as an independent prognosis factor for OVCA patients.


Subject(s)
Calcium-Binding Proteins/metabolism , GTPase-Activating Proteins/metabolism , Ovarian Neoplasms/metabolism , Animals , Carcinogenesis , Carrier Proteins , Cell Line, Tumor , Dimerization , Female , Gene Expression Regulation, Neoplastic , Glycolysis , HEK293 Cells , Humans , Membrane Proteins , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Ovarian Neoplasms/genetics , Prognosis , Protein Domains , Pyruvate Kinase/metabolism , Thyroid Hormones , Up-Regulation , Thyroid Hormone-Binding Proteins
20.
Front Pharmacol ; 9: 1295, 2018.
Article in English | MEDLINE | ID: mdl-30483132

ABSTRACT

Non-coding RNAs (ncRNAs) are defined as RNA molecules that do not encode proteins, but recent evidence has proven that peptides/proteins encoded by ncRNAs do indeed exist and usually contain less than 100 amino acids. These peptides/proteins play an important role in regulating tumor energy metabolism, epithelial to mesenchymal transition of cancer cells, the stability of the c-Myc oncoprotein, and the ubiquitination and degradation of proliferating cell nuclear antigen (PCNA). These peptides/proteins represent promising drug targets for fighting against tumor growth or biomarkers for predicting the prognosis of cancer patients. In this review, we summarize the characteristics of peptides/proteins that have recently been identified as putative ncRNA translation products and their outlook for small molecule peptide drugs, drug targets, and biomarkers.

SELECTION OF CITATIONS
SEARCH DETAIL