Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
PLoS Biol ; 21(12): e3002446, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38134227

ABSTRACT

Tumor metastasis is the major cause of breast cancer morbidity and mortality. It has been reported that the F-box protein FBXO3 functions as an E3 ubiquitin ligase in regulating various biological processes, including host autoimmune, antiviral innate immunity, and inflammatory response. However, the role of FBXO3 in tumor metastasis remains elusive. We have previously shown that ΔNp63α is a common inhibitory target in oncogene-induced cell motility and tumor metastasis. In this study, we show that FBXO3 plays a vital role in PI3K-mediated breast cancer metastasis independent of its E3 ligase activity and ΔNp63α in breast cancer cells and in mouse. FBXO3 can bind to and stabilize USP4, leading to Twist1 protein stabilization and increased breast cancer cell migration and tumor metastasis. Mechanistically, FBXO3 disrupts the interaction between USP4 and aspartyl aminopeptidase (DNPEP), thereby protecting USP4 from DNPEP-mediated degradation. Furthermore, p110αH1047R facilitates the phosphorylation and stabilization of FBXO3 in an ERK1-dependent manner. Knockdown of either FBXO3 or USP4 leads to significant inhibition of PI3K-induced breast cancer metastasis. Clinically, elevated expression of p110α/FBXO3/USP4/Twist1 is associated with poor overall survival (OS) and recurrence-free survival (RFS) of breast cancer patients. Taken together, this study reveals that the FBXO3-USP4-Twist1 axis is pivotal in PI3K-mediated breast tumor metastasis and that FBXO3/USP4 may be potential therapeutic targets for breast cancer treatment.


Subject(s)
Breast Neoplasms , Melanoma , Skin Neoplasms , Animals , Female , Humans , Mice , Breast Neoplasms/metabolism , Cell Line, Tumor , Phosphatidylinositol 3-Kinases/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitination
2.
Nucleic Acids Res ; 51(W1): W17-W24, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37207341

ABSTRACT

The utilization of high-throughput sequencing (HTS) for B-cell receptor (BCR) immune repertoire analysis has become widespread in the fields of adaptive immunity and antibody drug development. However, the sheer volume of sequences generated by these experiments presents a challenge in data processing. Specifically, multiple sequence alignment (MSA), a critical aspect of BCR analysis, remains inadequate for handling massive BCR sequencing data and lacks the ability to provide immunoglobulin-specific information. To address this gap, we introduce Abalign, a standalone program specifically designed for ultrafast MSA of BCR/antibody sequences. Benchmark tests demonstrate that Abalign achieves comparable or even better accuracy than state-of-the-art MSA tools, and shows remarkable advantages in terms of speed and memory consumption, reducing the time required for high-throughput analysis from weeks to hours. In addition to its alignment capabilities, Abalign offers a broad range of BCR analysis features, including extracting BCRs, constructing lineage trees, assigning VJ genes, analyzing clonotypes, profiling mutations, and comparing BCR immune repertoires. With its user-friendly graphic interface, Abalign can be easily run on personal computers instead of computing clusters. Overall, Abalign is an easy-to-use and effective tool that enables researchers to analyze massive BCR/antibody sequences, leading to new discoveries in the field of immunoinformatics. The software is freely available at http://cao.labshare.cn/abalign/.


Subject(s)
Antibodies , Software , Sequence Alignment , Antibodies/genetics , Adaptive Immunity , High-Throughput Nucleotide Sequencing/methods , Receptors, Antigen, B-Cell/genetics
3.
Brief Bioinform ; 23(3)2022 05 13.
Article in English | MEDLINE | ID: mdl-35289358

ABSTRACT

Protein-ligand docking is an essential method in computer-aided drug design and structural bioinformatics. It can be used to identify active compounds and reveal molecular mechanisms of biological processes. A successful docking usually requires thorough conformation sampling and scoring, which are computationally expensive and difficult. Recent studies demonstrated that it can be beneficial to docking with the guidance of existing similar co-crystal structures. In this work, we developed a protein-ligand docking method, named FitDock, which fits initial conformation to the given template using a hierarchical multi-feature alignment approach, subsequently explores the possible conformations and finally outputs refined docking poses. In our comprehensive benchmark tests, FitDock showed 40%-60% improvement in terms of docking success rate and an order of magnitude faster over popular docking methods, if template structures exist (> 0.5 ligand similarity). FitDock has been implemented in a user-friendly program, which could serve as a convenient tool for drug design and molecular mechanism exploration. It is now freely available for academic users at http://cao.labshare.cn/fitdock/.


Subject(s)
Drug Design , Proteins , Binding Sites , Ligands , Molecular Docking Simulation , Protein Binding , Protein Conformation , Proteins/chemistry
4.
PLoS Biol ; 19(2): e3001113, 2021 02.
Article in English | MEDLINE | ID: mdl-33626035

ABSTRACT

Transforming growth factor-ß (TGF-ß) signaling plays a critical role in promoting epithelial-to-mesenchymal transition (EMT), cell migration, invasion, and tumor metastasis. ΔNp63α, the major isoform of p63 protein expressed in epithelial cells, is a key transcriptional regulator of cell adhesion program and functions as a critical metastasis suppressor. It has been documented that the expression of ΔNp63α is tightly controlled by oncogenic signaling and is frequently reduced in advanced cancers. However, whether TGF-ß signaling regulates ΔNp63α expression in promoting metastasis is largely unclear. In this study, we demonstrate that activation of TGF-ß signaling leads to stabilization of E3 ubiquitin ligase FBXO3, which, in turn, targets ΔNp63α for proteasomal degradation in a Smad-independent but Erk-dependent manner. Knockdown of FBXO3 or restoration of ΔNp63α expression effectively rescues TGF-ß-induced EMT, cell motility, and tumor metastasis in vitro and in vivo. Furthermore, clinical analyses reveal a significant correlation among TGF-ß receptor I (TßRI), FBXO3, and p63 protein expression and that high expression of TßRI/FBXO3 and low expression of p63 are associated with poor recurrence-free survival (RFS). Together, these results demonstrate that FBXO3 facilitates ΔNp63α degradation to empower TGF-ß signaling in promoting tumor metastasis and that the TßRI-FBXO3-ΔNp63α axis is critically important in breast cancer development and clinical prognosis. This study suggests that FBXO3 may be a potential therapeutic target for advanced breast cancer treatment.


Subject(s)
Breast Neoplasms/pathology , Signal Transduction , Transforming Growth Factor beta/metabolism , Ubiquitin-Protein Ligases/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , HEK293 Cells , HaCaT Cells , Humans , Neoplasm Metastasis/pathology , Protein Isoforms , Tumor Suppressor Proteins/metabolism
5.
Nucleic Acids Res ; 50(W1): W159-W164, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35609983

ABSTRACT

Protein-ligand blind docking is a powerful method for exploring the binding sites of receptors and the corresponding binding poses of ligands. It has seen wide applications in pharmaceutical and biological researches. Previously, we proposed a blind docking server, CB-Dock, which has been under heavy use (over 200 submissions per day) by researchers worldwide since 2019. Here, we substantially improved the docking method by combining CB-Dock with our template-based docking engine to enhance the accuracy in binding site identification and binding pose prediction. In the benchmark tests, it yielded the success rate of ∼85% for binding pose prediction (RMSD < 2.0 Å), which outperformed original CB-Dock and most popular blind docking tools. This updated docking server, named CB-Dock2, reconfigured the input and output web interfaces, together with a highly automatic docking pipeline, making it a particularly efficient and easy-to-use tool for the bioinformatics and cheminformatics communities. The web server is freely available at https://cadd.labshare.cn/cb-dock2/.


Subject(s)
Algorithms , Proteins , Binding Sites , Ligands , Molecular Docking Simulation , Protein Binding , Proteins/chemistry , Software , GTPase-Activating Proteins/chemistry , Guanine Nucleotide Exchange Factors/chemistry
6.
J Cell Sci ; 134(23)2021 12 01.
Article in English | MEDLINE | ID: mdl-34761265

ABSTRACT

Protein abnormalities can accelerate aging causing protein misfolding diseases, and various adaptive responses have evolved to relieve proteotoxicity. To trigger these responses, cells must detect the buildup of aberrant proteins. Previously we demonstrated that the Hsp70-Bag3 (HB) complex senses the accumulation of defective ribosomal products, stimulating signaling pathway proteins, such as stress kinases or the Hippo pathway kinase LATS1. Here, we studied how Bag3 regulates the ability for LATS1 to regulate its key downstream target YAP (also known as YAP1). In naïve cells, Bag3 recruited a complex of LATS1, YAP and the scaffold AmotL2, which links LATS1 and YAP. Upon inhibition of the proteasome, AmotL2 dissociated from Bag3, which prevented phosphorylation of YAP by LATS1, and led to consequent nuclear YAP localization together with Bag3. Mutations in Bag3 that enhanced its translocation into nucleus also facilitated nuclear translocation of YAP. Interestingly, Bag3 also controlled YAP nuclear localization in response to cell density, indicating broader roles beyond proteotoxic signaling responses for Bag3 in the regulation of YAP. These data implicate Bag3 as a regulator of Hippo pathway signaling, and suggest mechanisms by which proteotoxic stress signals are propagated.


Subject(s)
Adaptor Proteins, Signal Transducing , Hippo Signaling Pathway , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Proliferation , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Brief Bioinform ; 22(4)2021 07 20.
Article in English | MEDLINE | ID: mdl-33313714

ABSTRACT

Identifying the types of body fluids left at the crime scene can be essential to reconstructing the crime scene and inferring criminal behavior. MicroRNA (miRNA) molecule extracted from the trace of body fluids is one of the most promising biomarkers for the identification due to its high expression, extreme stability and tissue specificity. However, the detection of miRNA markers is not the answer to a yes-no question but the probability of an assumption. Therefore, it is a crucial task to develop complicated methods combining multi-miRNAs as well as computational algorithms to achieve the goal. In this study, we systematically analyzed the expression of 10 most probable body fluid-specific miRNA markers (miR-451a, miR-205-5p, miR-203a-3p, miR-214-3p, miR-144-3p, miR-144-5p, miR-654-5p, miR-888-5p, miR-891a-5p and miR-124-3p) in 605 body fluids-related samples, including peripheral blood, menstrual blood, saliva, semen and vaginal secretion. We introduced the kernel density estimation (KDE) method and six well-established methods to classify the body fluids in order to find the most optimal combinations of miRNA markers as well as the corresponding classifying method. The results show that the combination of miR-451a, miR-891a-5p, miR-144-5p and miR-203a-3p together with KDE can achieve the most accurate and robust performance according to the cross-validation, independent tests and random perturbation tests. This systematic analysis suggests a reference scheme for the identification of body fluids in an accurate and stable manner.


Subject(s)
Body Fluids , Forensic Genetics , Genetic Markers , MicroRNAs/genetics , Real-Time Polymerase Chain Reaction , Adult , Algorithms , Female , Humans , Male
8.
Acta Pharmacol Sin ; 44(4): 888-896, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36216900

ABSTRACT

Computationally identifying new targets for existing drugs has drawn much attention in drug repurposing due to its advantages over de novo drugs, including low risk, low costs, and rapid pace. To facilitate the drug repurposing computation, we constructed an automated and parameter-free virtual screening server, namely DrugRep, which performed molecular 3D structure construction, binding pocket prediction, docking, similarity comparison and binding affinity screening in a fully automatic manner. DrugRep repurposed drugs not only by receptor-based screening but also by ligand-based screening. The former automatically detected possible binding pockets of the receptor with our cavity detection approach, and then performed batch docking over drugs with a widespread docking program, AutoDock Vina. The latter explored drugs using seven well-established similarity measuring tools, including our recently developed ligand-similarity-based methods LigMate and FitDock. DrugRep utilized easy-to-use graphic interfaces for the user operation, and offered interactive predictions with state-of-the-art accuracy. We expect that this freely available online drug repurposing tool could be beneficial to the drug discovery community. The web site is http://cao.labshare.cn/drugrep/ .


Subject(s)
Databases, Pharmaceutical , Drug Repositioning , Binding Sites , Drug Discovery/instrumentation , Drug Discovery/methods , Drug Repositioning/instrumentation , Ligands , Molecular Docking Simulation
9.
Proc Natl Acad Sci U S A ; 117(14): 8013-8021, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32193335

ABSTRACT

AMP-activated protein kinase (AMPK) functions as an energy sensor and is pivotal in maintaining cellular metabolic homeostasis. Numerous studies have shown that down-regulation of AMPK kinase activity or protein stability not only lead to abnormality of metabolism but also contribute to tumor development. However, whether transcription regulation of AMPK plays a critical role in cancer metastasis remains unknown. In this study, we demonstrate that AMPKα1 expression is down-regulated in advanced human breast cancer and is associated with poor clinical outcomes. Transcription of AMPKα1 is inhibited on activation of PI3K and HER2 through ΔNp63α. Ablation of AMPKα1 expression or inhibition of AMPK kinase activity leads to disruption of E-cadherin-mediated cell-cell adhesion in vitro and increased tumor metastasis in vivo. Furthermore, restoration of AMPKα1 expression significantly rescues PI3K/HER2-induced disruption of cell-cell adhesion, cell invasion, and cancer metastasis. Together, these results demonstrate that the transcription control is another layer of AMPK regulation and suggest a critical role for AMPK in regulating cell-cell adhesion and cancer metastasis.


Subject(s)
AMP-Activated Protein Kinases/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Breast/pathology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line, Tumor , Chromones/pharmacology , Disease-Free Survival , Down-Regulation , Female , Gene Knockdown Techniques , Humans , Lapatinib/pharmacology , Mice , Morpholines/pharmacology , Neoplasm Staging , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Prognosis , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Tissue Array Analysis , Transcription, Genetic/drug effects , Transcriptional Activation , Xenograft Model Antitumor Assays
11.
Nucleic Acids Res ; 48(D1): D797-D806, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31701131

ABSTRACT

Comprehensive genomic analyses of cancers have revealed substantial intrapatient molecular heterogeneities that may explain some instances of drug resistance and treatment failures. Examination of the clonal composition of an individual tumor and its evolution through disease progression and treatment may enable identification of precise therapeutic targets for drug design. Multi-region and single-cell sequencing are powerful tools that can be used to capture intratumor heterogeneity. Here, we present a database we've named CancerTracer (http://cailab.labshare.cn/cancertracer): a manually curated database designed to track and characterize the evolutionary trajectories of tumor growth in individual patients. We collected over 6000 tumor samples from 1548 patients corresponding to 45 different types of cancer. Patient-specific tumor phylogenetic trees were constructed based on somatic mutations or copy number alterations identified in multiple biopsies. Using the structured heterogeneity data, researchers can identify common driver events shared by all tumor regions, and the heterogeneous somatic events present in different regions of a tumor of interest. The database can also be used to investigate the phylogenetic relationships between primary and metastatic tumors. It is our hope that CancerTracer will significantly improve our understanding of the evolutionary histories of tumors, and may facilitate the identification of predictive biomarkers for personalized cancer therapies.


Subject(s)
Databases, Factual , Disease Progression , Neoplasms , Biomarkers, Tumor/genetics , Genetic Heterogeneity , Humans , Mutation , Neoplasms/classification , Neoplasms/genetics , Neoplasms/pathology , Phenotype , Phylogeny
12.
Acta Pharmacol Sin ; 41(1): 138-144, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31263275

ABSTRACT

As the number of elucidated protein structures is rapidly increasing, the growing data call for methods to efficiently exploit the structural information for biological and pharmaceutical purposes. Given the three-dimensional (3D) structure of a protein and a ligand, predicting their binding sites and affinity are a key task for computer-aided drug discovery. To address this task, a variety of docking tools have been developed. Most of them focus on docking in the preset binding sites given by users. To automatically predict binding modes without information about binding sites, we developed a user-friendly blind docking web server, named CB-Dock, which predicts binding sites of a given protein and calculates the centers and sizes with a novel curvature-based cavity detection approach, and performs docking with a popular docking program, Autodock Vina. This method was carefully optimized and achieved ~70% success rate for the top-ranking poses whose root mean square deviation (RMSD) were within 2 Å from the X-ray pose, which outperformed the state-of-the-art blind docking tools in our benchmark tests. CB-Dock offers an interactive 3D visualization of results, and is freely available at http://cao.labshare.cn/cb-dock/.


Subject(s)
Internet , Molecular Docking Simulation , Proteins/chemistry , Software , Algorithms , Binding Sites , Databases, Protein , Drug Design , Ligands
13.
Proc Natl Acad Sci U S A ; 114(20): E3964-E3973, 2017 05 16.
Article in English | MEDLINE | ID: mdl-28468801

ABSTRACT

Activation of phosphatidylinositol 3 kinase (PI3K), Ras, and Her2 signaling plays a critical role in cancer development. Hotspot constitutive activating mutations in oncogenes, such as PIK3CA encoding the p110α catalytic subunit or RAS, as well as overexpression of Her2, are frequently found in human tumors and cancers. It has been well established that activation of these oncogenes profoundly promotes tumor metastasis, whereas decreased expression of ΔNp63α, the major protein isoform of the p53-related p63 expressed in epithelial cells, has been associated with cancer metastasis. In this study, we demonstrate that hotspot oncogenic mutations on PIK3CA and RAS, including p110αH1047R, K-RasG12V, and H-RasG12V, as well as activation of Her2, all led to suppression of ΔNp63α expression via Akt-fork-head transcription factor 3a (Akt-FOXO3a) signaling, resulting in increased cell motility and tumor metastasis. Expression of ΔNp63α effectively reversed p110αH1047R-, K-RasG12V-, H-RasG12V-, or Her2-induced cell motility in vitro and tumor metastasis in mouse models. We show that ΔNp63α was a direct FOXO3a transcriptional target and that expression of FOXO3a and ΔNp63α was correlated in human cancer biopsy samples. Together, these results demonstrate that ΔNp63α is a common inhibitory target of oncogenic PI3K, Ras, and Her2, and that ΔNp63α may function as a critical integrator of oncogenic signaling in cancer metastasis.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Female , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genes, ras/genetics , Humans , Mice , Mutation , Neoplasm Metastasis/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction
14.
Biochem Biophys Res Commun ; 509(3): 784-789, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30635119

ABSTRACT

p53-related p63 plays a critical role in regulation of cell proliferation, survival and cell differentiation. Dysregulation of p63 functions results in a disruption of a variety of normal biological processes, including stem cell biology, embryonic development, aging and tumorigenesis. ΔNp63α, a predominantly expressed p63 protein isoform in epithelial cells, plays a crucial role in regulation of cell cycle progression and cell growth. p38 MAP kinases (p38MAPK) are the members of mitogen-activated protein kinases family and are critical in regulation of cell survival in response to stress signals. In this study, we show that ectopic expression of ΔNp63α inhibited phosphorylation of p38MAPK. Acute knockdown of p63 led to a significant upregulation of p38MAPK phosphorylation, resulting in increased p21cip1/waf1 expression, reduced phosphorylation of retinoblastoma protein (RB), cell cycle G1 arrest and cell growth retardation. Restoration of ΔNp63α expression reversed cell cycle arrest and growth inhibition induced by p63 ablation. Pharmacological inhibition of p38MAPK significantly suppressed ΔNp63α ablation-induced cell cycle G1/S arrest. In addition, MAP Kinase Phosphatase 3 (MKP3) was responsible for ΔNp63α-mediated regulation of p38MAPK phosphorylation. Together, these results suggest that ΔNp63α-MPK3-p38MAPK signaling pathway plays an important role in cell cycle progression and cell growth.


Subject(s)
Cell Proliferation , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Cycle , Cell Cycle Checkpoints , Cell Line, Tumor , Dual Specificity Phosphatase 6/metabolism , Enzyme Activation , Humans , Phosphorylation , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
15.
Cell Mol Life Sci ; 75(6): 965-973, 2018 03.
Article in English | MEDLINE | ID: mdl-28975366

ABSTRACT

As a member of tumor suppressor p53 family, p63, a gene encoding versatile protein variant, has been documented to correlate with cancer formation and progression, though it is rarely mutated in cancer patients. However, it has long been controversial on whether p63 is an oncogene or a tumor suppressor. Here, we comprehensively reviewed reports on roles of p63 in development, tumorigenesis and tumor progression. According to data from molecular cell biology, genetic models and clinic research, we conclude that p63 may act as either an oncogene or a tumor suppressor gene in different scenarios: TA isoforms of p63 gene are generally tumor-suppressive through repressing cell proliferation, survival and metastasis; ΔN isoforms, however, may initiate tumorigenesis via promoting cell proliferation and survival, but inhibit tumor metastasis and progression; effects of p63 on tumor formation and progression depend on the context of the whole p53 family, and either amplification or loss of p63 gene locus can break the balance to cause tumorigenesis.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Cell Survival , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Neoplasms/metabolism , Neoplasms/pathology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
16.
Int J Mol Sci ; 20(9)2019 May 06.
Article in English | MEDLINE | ID: mdl-31064130

ABSTRACT

HER2 (human epidermal growth factor receptor 2) activation is critical in breast cancer development. HER2 promotes cell proliferation, angiogenesis, survival, and metastasis by activation of PI3K/Akt, Ras/MEK/ERK, and JAK/STAT pathways. However, beyond these signaling molecules, the key proteins underlining HER2-mediated metastasis remain elusive. ATF4 (Activating transcription factor 4), a critical regulator in unfolded protein response (UPR), is implicated in cell migration and tumor metastasis. In this study, we demonstrate that HER2 upregulated ATF4 expression at both mRNA and protein levels, resulting in cell migration increased. In addition, ATF4 upregulated ZEB1 (Zinc finger E-box-binding homeobox 1) and suppressed E-cadherin expression resulting in promoting cell migration. Restoration of E-cadherin expression effectively inhibited HER2- or ATF4-mediated cell migration. In addition, upregulated expression of ATF4 was found in HER2-positive breast cancer specimens. Together, this study demonstrates that ATF4-ZEB1 is important for HER2-mediated cell migration and suggests that ATF4-ZEB1 may be potential therapeutic targets for breast cancer metastasis.


Subject(s)
Activating Transcription Factor 4/genetics , Breast Neoplasms/metabolism , Cadherins/genetics , Cell Movement , Receptor, ErbB-2/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Activating Transcription Factor 4/metabolism , Breast Neoplasms/genetics , Cadherins/metabolism , Cell Line , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Receptor, ErbB-2/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
17.
Int J Mol Sci ; 20(5)2019 Mar 07.
Article in English | MEDLINE | ID: mdl-30866414

ABSTRACT

Cell⁻cell adhesion plays an important role in regulation of cell proliferation, migration, survival, and drug sensitivity. Metformin, a first line drug for type 2 diabetes, has been shown to possess anti-cancer activities. However, whether cell⁻cell adhesion affects metformin anti-cancer activity is unknown. In this study, Microscopic and FACS analyses showed that metformin induced cancer cell⁻cell adhesion exemplified by cell aggregation and anoikis under glucose restriction. Furthermore, western blot and QPCR analyses revealed that metformin dramatically upregulated integrin ß1 expression. Silencing of integrin ß1 significantly disrupted cell aggregation and reduced anoikis induced by metformin. Moreover, we showed that p53 family member ΔNp63α transcriptionally suppressed integrin ß1 expression and is responsible for metformin-mediated upregulation of integrin ß1. In summary, this study reveals a novel mechanism for metformin anticancer activity and demonstrates that cell⁻cell adhesion mediated by integrin ß1 plays a critical role in metformin-induced anoikis.


Subject(s)
Glucose/pharmacology , Integrin beta1/genetics , Integrin beta1/metabolism , Metformin/pharmacology , Neoplasms/metabolism , Anoikis , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , HEK293 Cells , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation
18.
J Biol Chem ; 292(13): 5253-5261, 2017 Mar 31.
Article in English | MEDLINE | ID: mdl-28193839

ABSTRACT

The blood glucose modifier metformin is used to treat type II diabetes and has also been shown to possess anticancer activities. Recent studies indicate that glucose deprivation can greatly enhance metformin-mediated inhibition of cell viability, but the molecular mechanism involved in this inhibition is unclear. In this study, we report that, under glucose deprivation, metformin inhibited expression of ΔNp63α, a p53 family member involved in cell adhesion pathways, resulting in disruption of cell matrix adhesion and subsequent apoptosis in human squamous carcinoma cells. We further show that metformin promoted ΔNp63α protein instability independent of AMP-activated protein kinase and that WWP1, an E3 ligase of ΔNp63α, was involved in metformin-mediated down-regulation of ΔNp63α levels. In addition, we demonstrate that a combination of metformin and the glycolysis inhibitor 2-deoxy-d-glucose significantly inhibited ΔNp63α expression and also suppressed xenographic tumor growth in vivo In summary, this study reveals a new mechanism for metformin-mediated anticancer activity and suggests a new strategy for treating human squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Metformin/pharmacology , Transcription Factors/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors , AMP-Activated Protein Kinases , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Survival/drug effects , Deoxyglucose/pharmacology , Drug Therapy, Combination , Heterografts , Humans , Metformin/therapeutic use , Mice , Protein Stability/drug effects
19.
BMC Genomics ; 19(1): 138, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29433427

ABSTRACT

BACKGROUND: The integration of DNA methylation and copy number alteration data promises to provide valuable insight into the underlying molecular mechanisms responsible for cancer initiation and progression. However, the generation and processing of these datasets are costly and time-consuming if carried out separately. The Illumina Infinium HumanMethylation450 BeadChip, initially designed for the evaluation of DNA methylation levels, allows copy number variant calling using bioinformatics tools. RESULTS: A substantial amount of Infinium HumanMethylation450 data across various cancer types has been accumulated in recent years and is a valuable resource for large-scale data analysis. Here we present MethCNA, a comprehensive database for genomic and epigenomic data integration in human cancer. In the current release, MethCNA contains about 10,000 tumor samples representing 37 cancer types. All raw array data were collected from The Cancer Genome Atlas and NCBI Gene Expression Omnibus database and analyzed using a pipeline that integrated multiple computational resources and tools. The normalized copy number aberration data and DNA methylation alterations were obtained. We provide a user-friendly web-interface for data mining and visualization. CONCLUSIONS: The Illumina Infinium HumanMethylation450 BeadChip enables the interrogation and integration of both genomic and epigenomic data from exactly the same DNA specimen, and thus can aid in distinguishing driver from passenger mutations in cancer. We expect MethCNA will enable researchers to explore DNA methylation and copy number alteration patterns, identify key oncogenic drivers in cancer, and assist in the development of targeted therapies. MethCNA is publicly available online at http://cgma.scu.edu.cn/MethCNA .


Subject(s)
Computational Biology/methods , Databases, Genetic , Epigenomics/methods , Genomics/methods , Neoplasms/genetics , DNA Copy Number Variations , DNA Methylation , DNA, Neoplasm/genetics , Gene Expression Profiling/methods , Humans , Internet , Neoplasms/classification , Reproducibility of Results
20.
Appl Opt ; 57(25): 7242-7248, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30182985

ABSTRACT

AlGaInAs/InP waveguide-coupled deformed hexagonal resonator microlasers with enhanced mode quality (Q)-factors are demonstrated for realizing single-transverse-mode operation. A circular hole is introduced to the center of the hexagonal resonators with rounded corners to enhance the mode Q-factors and suppress high-order transverse modes simultaneously. Single-mode lasing with side-mode suppression ratios up to 40 dB is obtained for the 10-µm-sidelength hexagonal microlasers with a center hole. All the lasing spectra demonstrate pure single-transverse-mode properties within the whole tuning range of injection current, and mode hopping with one, two, and three longitudinal-mode intervals is observed due to the mode Q-factor modification by the center holes. To further reduce the device size and threshold current, the deformed hexagonal resonator microlasers with the flat sides replaced by circular arcs are analyzed and demonstrated experimentally. The Q-factors of the fundamental transverse modes can be enhanced by two orders of magnitude due to the convergence effect of the circular sides by optimizing the deformation amplitude, while the single-transverse-mode property is still maintained. A threshold current of 2.4 mA is realized for a circular-side hexagonal microlaser with the side length of 8.5 µm and the deformation amplitude of 0.55 µm.

SELECTION OF CITATIONS
SEARCH DETAIL