Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Br J Cancer ; 118(4): 522-533, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29348487

ABSTRACT

BACKGROUND: The therapeutic resistance to ionising radiation (IR) and anti-angiogenesis mainly impair the prognosis of patients with glioblastoma. The primary and secondary MET aberrant activation is one crucial factor for these resistances. The kringle 1 domain of hepatocyte growth factor (HGFK1), an angiogenic inhibitor, contains a high-affinity binding domain of MET; however, its effects on glioblastoma remain elusive. METHODS: We formed the nanoparticles consisting of a folate receptor-targeted nanoparticle-mediated HGFK1 gene (H1/pHGFK1) and studied its anti-tumoural and radiosensitive activities in both subcutaneous and orthotopic human glioma cell-xenografted mouse models. We then elucidated its molecular mechanisms in human glioblastoma cell lines in vitro. RESULTS: We demonstrated for the first time that peritumoural injection of H1/pHGFK1 nanoparticles significantly inhibited tumour growth and prolonged survival in tumour-bearing mice, as well as enhanced the anti-tumoural efficacies of IR in vivo by reducing Ki-67 expression, enhancing TUNEL staining-indicated apoptotic indexes, reducing microvascular intensity and reversing IR-induced MET overexpression in tumour tissues. Furthermore, we showed that HGFK1 suppressed the proliferation and induced cell apoptosis and enhanced sensitivity to IR in glioblastoma cell lines, mainly by suppressing the activities of MET receptor, down-regulating ATM-Chk2 axis but up-regulating Chk1. CONCLUSIONS: H1/pHGFK1 exerts anti-tumoural and radiosensitive activities mainly through the inhibition and reversal of IR-induced MET and ATM-Chk2 axis activities in glioblastoma. H1/pHGFK1 nanoparticles are a potential radiosensitiser and angiogenic inhibitor for glioblastoma treatment.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Hepatocyte Growth Factor/genetics , Plasmids/administration & dosage , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Radiation-Sensitizing Agents/administration & dosage , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/genetics , Hepatocyte Growth Factor/chemistry , Humans , Kringles , Mice , Nanoparticles/administration & dosage , Plasmids/genetics , Radiation-Sensitizing Agents/pharmacology , Xenograft Model Antitumor Assays
2.
Cancer Res ; 77(8): 1905-1917, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28196904

ABSTRACT

Mechanisms underlying the propensity of latent lung adenocarcinoma (LUAD) to relapse are poorly understood. In this study, we show how differential expression of a network of extracellular matrix (ECM) molecules and their interacting proteins contributes to risk of relapse in distinct LUAD subtypes. Overexpression of the hyaluronan receptor HMMR in primary LUAD was associated with an inflammatory molecular signature and poor prognosis. Attenuating HMMR in LUAD cells diminished their ability to initiate lung tumors and distant metastases. HMMR upregulation was not required for dissemination in vivo, but enhanced ECM-mediated signaling, LUAD cell survival, and micrometastasis expansion in hyaluronan-rich microenvironments in the lung and brain metastatic niches. Our findings reveal an important mechanism by which disseminated cancer cells can coopt the inflammatory ECM to persist, leading to brain metastatic outgrowths. Cancer Res; 77(8); 1905-17. ©2017 AACR.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Extracellular Matrix Proteins/biosynthesis , Hyaluronan Receptors/biosynthesis , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Cell Line, Tumor , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix Proteins/genetics , Gene Expression , Humans , Hyaluronan Receptors/genetics , Lung Neoplasms/genetics , Male , Mice , Mice, Nude , Neoplasm Micrometastasis , Transcriptome , Tumor Microenvironment
3.
Nucleic Acids Res ; 44(19): e149, 2016 Nov 02.
Article in English | MEDLINE | ID: mdl-27458201

ABSTRACT

The CRISPR/Cas9 system is a powerful genome editing tool and has been widely used for biomedical research. However, many challenges, such as off-target effects and lack of easy solutions for multiplex targeting, are still limiting its applications. To overcome these challenges, we first developed a highly efficient doxycycline-inducible Cas9-EGFP vector. This vector allowed us to track the cells for uniform temporal control and efficient gene disruption, even in a polyclonal setting. Furthermore, the inducible CRISPR/Cas9 system dramatically decreased off-target effects with a pulse exposure of the genome to the Cas9/sgRNA complex. To target multiple genes simultaneously, we established simple one-step cloning approaches for expression of multiple sgRNAs with improved vectors. By combining our inducible and multiplex genome editing approaches, we were able to simultaneously delete Lysine Demethylase (KDM) 5A, 5B and 5C efficiently in vitro and in vivo This user friendly and highly efficient toolbox provides a solution for easy genome editing with tight temporal control, minimal off-target effects and multiplex targeting.


Subject(s)
CRISPR-Cas Systems , Gene Targeting , Bacterial Proteins/metabolism , CRISPR-Associated Protein 9 , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , Endonucleases/metabolism , Gene Expression , Gene Knockout Techniques , Gene Order , Gene Silencing , Gene Targeting/methods , Gene Targeting/standards , Genes, Reporter , Genetic Vectors/genetics , Humans , Promoter Regions, Genetic , RNA, Guide, Kinetoplastida , Retinoblastoma-Binding Protein 2/deficiency
4.
Cell Rep ; 6(5): 868-77, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24582965

ABSTRACT

Metastasis is a major clinical challenge for cancer treatment. Emerging evidence suggests that aberrant epigenetic modifications contribute significantly to tumor formation and progression. However, the drivers and roles of such epigenetic changes in tumor metastasis are still poorly understood. Using bioinformatic analysis of human breast cancer gene-expression data sets, we identified histone demethylase RBP2 as a putative mediator of metastatic progression. By using both human breast cancer cells and genetically engineered mice, we demonstrated that RBP2 is critical for breast cancer metastasis to the lung in multiple in vivo models. Mechanistically, RBP2 promotes metastasis as a pleiotropic positive regulator of many metastasis genes, including TNC. In addition, RBP2 loss suppresses tumor formation in MMTV-neu transgenic mice. These results suggest that therapeutic targeting of RBP2 is a potential strategy for inhibition of tumor progression and metastasis.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Histone Demethylases/metabolism , Retinol-Binding Proteins, Cellular/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Histone Demethylases/genetics , Humans , Male , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplasm Metastasis , Retinol-Binding Proteins, Cellular/genetics , Transfection
5.
Cancer Cell ; 23(6): 725-38, 2013 Jun 10.
Article in English | MEDLINE | ID: mdl-23707782

ABSTRACT

Molecular programs that mediate normal cell differentiation are required for oncogenesis and tumor cell survival in certain cancers. How cell-lineage-restricted genes specifically influence metastasis is poorly defined. In lung cancers, we uncovered a transcriptional program that is preferentially associated with distal airway epithelial differentiation and lung adenocarcinoma (ADC) progression. This program is regulated in part by the lineage transcription factors GATA6 and HOPX. These factors can cooperatively limit the metastatic competence of ADC cells, by modulating overlapping alveolar differentiation and invasogenic target genes. Thus, GATA6 and HOPX are critical nodes in a lineage-selective pathway that directly links effectors of airway epithelial specification to the inhibition of metastasis in the lung ADC subtype.


Subject(s)
Adenocarcinoma/pathology , GATA6 Transcription Factor/physiology , Homeodomain Proteins/physiology , Lung Neoplasms/pathology , Neoplasm Metastasis/pathology , Tumor Suppressor Proteins/physiology , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Cluster Analysis , Epithelium/pathology , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lung Neoplasms/genetics , Neoplasm Invasiveness , Neoplasm Metastasis/genetics , Pulmonary Alveoli/cytology , Pulmonary Alveoli/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
6.
J Biol Chem ; 287(13): 9962-9971, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22253443

ABSTRACT

miR-124 is a brain-enriched microRNA that plays a crucial role in neural development and has been shown to be down-regulated in glioma and medulloblastoma, suggesting its possible involvement in brain tumor progression. Here, we show that miR-124 is down-regulated in a panel of different grades of glioma tissues and in all of the human glioma cell lines we examined. By integrated bioinformatics analysis and experimental confirmation, we identified SNAI2, which is often up-regulated in glioma, as a direct functional target of miR-124. Because SNAI2 has been shown to regulate stem cell functions, we examined the roles of miR-124 and SNAI2 in glioma cell stem-like traits. The results showed that overexpression of miR-124 and knockdown of SNAI2 reduced neurosphere formation, CD133(+) cell subpopulation, and stem cell marker (BMI1, Nanog, and Nestin) expression, and these effects could be rescued by re-expression of SNAI2. Furthermore, enhanced miR-124 expression significantly inhibited glioma cell invasion in vitro. Finally, stable overexpression of miR-124 and knockdown of SNAI2 inhibited the tumorigenicity and invasion of glioma cells in vivo. These findings reveal, for the first time, that the tumor suppressor activity of miR-124 could be partly due to its inhibitory effects on glioma stem-like traits and invasiveness through SNAI2.


Subject(s)
Antigens, Differentiation/metabolism , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Glioma/metabolism , MicroRNAs/biosynthesis , Neoplastic Stem Cells/metabolism , Animals , Antigens, Differentiation/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms , Cell Line, Tumor , Down-Regulation/genetics , Glioma/genetics , Glioma/pathology , Humans , Mice , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Snail Family Transcription Factors , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Up-Regulation/genetics
7.
Discov Med ; 12(65): 329-40, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22031670

ABSTRACT

Cancer has long been compared to the aberrant development of human tissues. It was in the mid-19th century writings of Rudolf Virchow and Joseph Recamier that malignant tissue was first proposed to originate from embryonal cells. More contemporary perspectives on malignant progression are founded on the tenant that tumors emerge from somatic tissues. Yet examples linking the biological properties of cancer to developmental processes, both aberrant and normal, abound. In this review, we will discuss how the developmental lineage of tumor cells can influence the course of cancer metastasis. As new molecular mechanisms that control cell fate in various tissues are being rapidly uncovered, understanding how these well orchestrated programs can be subverted in human diseases should provide intriguing avenues for fundamental biological discoveries and new therapeutic opportunities in cancer.


Subject(s)
Cell Lineage/physiology , Neoplasms/pathology , Animals , Disease Progression , Humans , Neoplasms/complications
8.
Comput Biol Chem ; 34(4): 232-41, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20863765

ABSTRACT

A number of microRNAs (miRNAs) that are evolutionarily conserved not beyond primate lineage have been identified. These primate-specific miRNAs (ps-miRNAs) may attribute to the difference between high-level primates and non-primate mammals or lower vertebrates. Despite of their importance, the genome-wide miRNA conservation patterns and the properties of these ps-miRNAs are largely elusive. In this study, we developed a robust classification system to assess the conservation pattern of all human mature miRNAs across 44 vertebrate genomes. By this comparative genomic analysis, a novel set of 269 ps-miRNAs were identified. We found that many ps-miRNAs were enriched in chromosome 19 and X, forming two main clusters hereafter referred as C19MC and CXMC, respectively. When comparing the seed of ps-miRNAs themselves or with non-ps-miRNAs, more than one half ps-miRNAs sharing common seeds were belonged to C19MC, 9 of which retained a unique seed that had been reported to be enriched in human embryonic stem cells (hESCs) specific miRNAs. Moreover, the most abundant ps-miRNA common seed was possessed by miR-548 family. Most ps-miRNAs had very low expression in adult tissues, which may be attributed to temporal and spatial specific transcript regulation. The ps-miRNAs with relatively high expression were mainly belonged to C19MC and CXMC, and preferentially expressed in hESCs and reproductive system. Sequence anatomy revealed that C19MC ps-miRNAs were highly conserved but not beyond primates and of great sequence similarity. Gene Ontology and KEGG pathway enrichment analyses of predicted target genes indicated that C19MC ps-miRNAs were strongly associated with developmental processes and various cancers. In conclusion, ps-miRNAs may play critical roles in differentiation and growth regulation during early development, especially in maintaining the pluripotency of hESCs. Results from this study may help explaining the differences between primates and lower vertebrates at genetic level.


Subject(s)
Genome, Human , Genomics/methods , MicroRNAs/genetics , Animals , Cell Line , Chromosomes, Human, Pair 19 , Chromosomes, Human, X , Embryonic Stem Cells/metabolism , Evolution, Molecular , Humans , Neoplasms/genetics , Primates/genetics
9.
J Biol Chem ; 285(47): 36995-7004, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20826811

ABSTRACT

The emerging concept of generating cancer stem cells from epithelial-mesenchymal transition has attracted great interest; however, the factors and molecular mechanisms that govern this putative tumor-initiating process remain largely elusive. We report here that miR-200a not only regulates epithelial-mesenchymal transition but also stem-like transition in nasopharyngeal carcinoma cells. We first showed that stable knockdown of miR-200a promotes the transition of epithelium-like CNE-1 cells to the mesenchymal phenotype. More importantly, it also induced several stem cell-like traits, including CD133(+) side population, sphere formation capacity, in vivo tumorigenicity in nude mice, and stem cell marker expression. Consistently, stable overexpression of miR-200a switched mesenchyme-like C666-1 cells to the epithelial state, accompanied by a significant reduction of stem-like cell features. Furthermore, in vitro differentiation of the C666-1 tumor sphere resulted in diminished stem-like cell population and miR-200a induction. To investigate the molecular mechanism, we demonstrated that miR-200a controls epithelial-mesenchymal transition by targeting ZEB2, although it regulates the stem-like transition differentially and specifically by ß-catenin signaling. Our findings reveal for the first time the function of miR-200a in shifting nasopharyngeal carcinoma cell states via a reversible process coined as epithelial-mesenchymal to stem-like transition through differential and specific mechanisms.


Subject(s)
Epithelial-Mesenchymal Transition , Homeodomain Proteins/metabolism , MicroRNAs/physiology , Nasopharyngeal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Repressor Proteins/metabolism , beta Catenin/metabolism , Animals , Blotting, Western , Cell Differentiation , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Immunoenzyme Techniques , Mice , Mice, Inbred BALB C , Mice, Nude , Nasopharyngeal Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Zinc Finger E-box Binding Homeobox 2 , beta Catenin/antagonists & inhibitors , beta Catenin/genetics
10.
Biochem Biophys Res Commun ; 394(1): 18-23, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20167204

ABSTRACT

Makorin-2, consisting of four highly conserved C(3)H zinc fingers, a Cys-His motif and a C(3)HC(4) RING zinc finger domain, is a putative ribonucleoprotein. We have previously reported that Xenopus makorin-2 (mkrn2) is a neurogenesis inhibitor acting upstream of glycogen synthase kinase-3beta (GSK-3beta) in the phosphatidylinositol 3-kinase/Akt pathway. In an effort to identify the functional domains required for its anti-neurogenic activity, we designed and constructed a series of N- and C-terminal truncation mutants of mkrn2. Concurred with the full-length mkrn2, we showed that overexpression of one of the truncation mutants mkrn2(s)-7, which consists of only the third C(3)H zinc finger, Cys-His motif and C(3)HC(4) RING zinc finger, is essential and sufficient to produce the phenotypical dorso-posterior deficiencies and small-head/short-tail phenotype in tadpoles. In animal cap explant assay, we further demonstrated that mkrn2(s)-7 not only inhibits activin and retinoic acid-induced animal cap neuralization and the expression of a pan-neural marker neural cell adhesion molecule, but also induces GSK-3beta expression. These results collectively suggest that the third C(3)H zinc finger, Cys-His motif and C(3)HC(4) RING zinc finger are indispensable for the anti-neurogenic activity of mkrn2.


Subject(s)
Neurogenesis , Ribonucleoproteins/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Amino Acid Motifs/genetics , Animals , Conserved Sequence , Embryo, Nonmammalian/metabolism , Glycogen Synthase Kinase 3/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phylogeny , Protein Structure, Tertiary/genetics , Ribonucleoproteins/classification , Ribonucleoproteins/genetics , Xenopus Proteins/classification , Xenopus Proteins/genetics , Xenopus laevis/genetics , Xenopus laevis/metabolism , Zinc Fingers/genetics
11.
Biochem Biophys Res Commun ; 391(1): 535-41, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19931509

ABSTRACT

Nasopharyngeal carcinoma (NPC), a highly metastatic and invasive malignant tumor originating from the nasopharynx, is widely prevalent in Southeast Asia, the Middle East and North Africa. Although viral, dietary and genetic factors have been implicated in NPC, the molecular basis of its pathogenesis is not well defined. Based on a recent microRNA (miRNA) microarray study showing miR-200 downregulation in NPC, we further investigated the role of miR-200a in NPC carcinogenesis. We found that the endogenous miR-200a expression level increases with the degree of differentiation in a panel of NPC cell lines, namely undifferentiated C666-1, high-differentiated CNE-1, and low-differentiated CNE-2 and HNE1 cells. By a series of gain-of-function and loss-of-function studies, we showed that over-expression of miR-200a inhibits C666-1 cell growth, migration and invasion, whereas its knock-down stimulates these processes in CNE-1 cells. In addition, we further identified ZEB2 and CTNNB1 as the functional downstream targets of miR-200a. Interestingly, knock-down of ZEB2 solely impeded NPC cell migration and invasion, whereas CTNNB1 suppression only inhibited NPC cell growth, suggesting that the inhibitory effects of miR-200a on NPC cell growth, migration and invasion are mediated by distinct targets and pathways. Our results reveal the important role of miR-200a as a regulatory factor of NPC carcinogenesis and a potential candidate for miRNA-based therapy against NPC.


Subject(s)
Carcinoma/pathology , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , MicroRNAs/physiology , Nasopharyngeal Neoplasms/pathology , Repressor Proteins/genetics , beta Catenin/genetics , Base Sequence , Carcinoma/genetics , Carcinoma/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Down-Regulation , Humans , MicroRNAs/genetics , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Neoplasm Invasiveness , Zinc Finger E-box Binding Homeobox 2
12.
J Biol Chem ; 283(13): 8486-95, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18198183

ABSTRACT

Makorin-2 belongs to the makorin RING zinc finger gene family, which encodes putative ribonucleoproteins. Here we cloned the Xenopus makorin-2 (mkrn2) and characterized its function in Xenopus neurogenesis. Forced overexpression of mkrn2 produced tadpoles with dorso-posterior deficiencies and small-head/short-tail phenotype, whereas knockdown of mkrn2 by morpholino antisense oligonucleotides induced double axis in tadpoles. In Xenopus animal cap explant assay, mkrn2 inhibited activin, and retinoic acid induced animal cap neuralization, as evident from the suppression of a pan neural marker, neural cell adhesion molecule. Surprisingly, the anti-neurogenic activity of mkrn2 is independent of the two major neurogenesis signaling cascades, BMP-4 and Wnt8 pathways. Instead, mkrn2 works specifically through the phosphatidylinositol 3-kinase (PI3K) and Akt-mediated neurogenesis pathway. Overexpression of mkrn2 completely abrogated constitutively active PI3K- and Akt-induced, but not dominant negative glycogen synthase kinase-3beta (GSK-3beta)-induced, neural cell adhesion molecule expression, indicating that mkrn2 acts downstream of PI3K and Akt and upstream of GSK-3beta. Moreover, mkrn2 up-regulated the mRNA and protein levels of GSK-3beta. These results revealed for the first time the important role of mkrn2 as a new player in PI3K/Akt-mediated neurogenesis during Xenopus embryonic development.


Subject(s)
Cell Differentiation , Neurons/cytology , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribonucleoproteins/metabolism , Signal Transduction , Xenopus Proteins/metabolism , Aging/physiology , Animals , Brain/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Female , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Molecular Sequence Data , Neural Cell Adhesion Molecules/genetics , Oocytes/metabolism , Phenotype , RNA, Messenger/genetics , Ribonucleoproteins/genetics , Skin/metabolism , Wnt Proteins/metabolism , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , Xenopus laevis/growth & development , Xenopus laevis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL