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











Publication year range
1.
Sci Rep ; 7: 40714, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28102366

ABSTRACT

The effects of AURKA overexpression associated with poor clinical outcomes have been attributed to increased cell cycle progression and the development of genomic instability with aneuploidy. We used RNA interference to examine the effects of AURKA overexpression in human bladder cancer cells. Knockdown had minimal effects on cell proliferation but blocked tumor cell invasion. Whole genome mRNA expression profiling identified nicotinamide N-methyltransferase (NNMT) as a downstream target that was repressed by AURKA. Chromatin immunoprecipitation and NNMT promoter luciferase assays revealed that AURKA's effects on NNMT were caused by PAX3-mediated transcriptional repression and overexpression of NNMT blocked tumor cell invasion in vitro. Overexpression of AURKA and activation of its downstream pathway was enriched in the basal subtype in primary human tumors and was associated with poor clinical outcomes. We also show that the FISH test for the AURKA gene copy number in urine yielded a specificity of 79.7% (95% confidence interval [CI] = 74.2% to 84.1%), and a sensitivity of 79.6% (95% CI = 74.2% to 84.1%) with an AUC of 0.901 (95% CI = 0.872 to 0.928; P < 0.001). These results implicate AURKA as an effective biomarker for bladder cancer detection as well as therapeutic target especially for its basal type.


Subject(s)
Aurora Kinase A/genetics , Biomarkers, Tumor , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/genetics , Aurora Kinase A/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Progression , Early Detection of Cancer , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization, Fluorescence , Prognosis , Transcription, Genetic , Urinary Bladder Neoplasms/mortality
2.
Cancer Res ; 76(11): 3145-55, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27013197

ABSTRACT

Nuclear factor of activated T cell (NFAT1, NFATC2) is a transcription factor that binds and positively regulates IL2 expression during T-cell activation. NFAT1 has important roles in both innate and adaptive immune responses, but its involvement in cancer is not completely understood. We previously demonstrated that NFAT1 contributes to melanoma growth and metastasis by regulating the autotaxin gene (Enpp2). Here, we report a strong correlation between NFAT1 expression and metastatic potential in melanoma cell lines and tumor specimens. To elucidate the mechanisms underlying NFAT1 overexpression during melanoma progression, we conducted a microarray on a highly metastatic melanoma cell line in which NFAT1 expression was stably silenced. We identified and validated two downstream targets of NFAT1, IL8, and MMP3. Accordingly, NFAT1 depletion in metastatic melanoma cell lines was associated with reduced IL8 and MMP3 expression, whereas NFAT1 overexpression in a weakly metastatic cell line induced expression of these targets. Restoration of NFAT1 expression recovered IL8 and MMP3 expression levels back to baseline, indicating that both are direct targets of NFAT1. Moreover, in vivo studies demonstrated that NFAT1 and MMP3 promoted melanoma tumor growth and lung metastasis. Collectively, our findings assign a new role for NFAT1 in melanoma progression, underscoring the multifaceted functions that immunomodulatory factors may acquire in an unpredictable tumor microenvironment. Cancer Res; 76(11); 3145-55. ©2016 AACR.


Subject(s)
Interleukin-8/metabolism , Lung Neoplasms/secondary , Matrix Metalloproteinase 3/metabolism , Melanoma/pathology , NFATC Transcription Factors/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cell Proliferation , Female , Humans , Immunoenzyme Techniques , Interleukin-8/genetics , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Matrix Metalloproteinase 3/genetics , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , NFATC Transcription Factors/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Nat Cell Biol ; 17(3): 311-21, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25686251

ABSTRACT

Although recent studies have shown that adenosine-to-inosine (A-to-I) RNA editing occurs in microRNAs (miRNAs), its effects on tumour growth and metastasis are not well understood. We present evidence of CREB-mediated low expression of ADAR1 in metastatic melanoma cell lines and tumour specimens. Re-expression of ADAR1 resulted in the suppression of melanoma growth and metastasis in vivo. Consequently, we identified three miRNAs undergoing A-to-I editing in the weakly metastatic melanoma but not in strongly metastatic cell lines. One of these miRNAs, miR-455-5p, has two A-to-I RNA-editing sites. The biological function of edited miR-455-5p is different from that of the unedited form, as it recognizes a different set of genes. Indeed, wild-type miR-455-5p promotes melanoma metastasis through inhibition of the tumour suppressor gene CPEB1. Moreover, wild-type miR-455 enhances melanoma growth and metastasis in vivo, whereas the edited form inhibits these features. These results demonstrate a previously unrecognized role for RNA editing in melanoma progression.


Subject(s)
Adenosine/metabolism , Gene Expression Regulation, Neoplastic , Inosine/metabolism , Melanoma/genetics , RNA Editing , Skin Neoplasms/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Progression , Female , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Nude , MicroRNAs , Molecular Sequence Data , Neoplasm Metastasis , Neoplasm Transplantation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
4.
Pigment Cell Melanoma Res ; 27(1): 19-36, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24106873

ABSTRACT

Malignant melanoma is one of the most aggressive cancers and can disseminate from a relatively small primary tumor and metastasize to multiple sites, including the lung, liver, brain, bone, and lymph nodes. Elucidating the molecular and genetic changes that take place during the metastatic process has led to a better understanding of why melanoma is so metastatic. Herein, we describe the unique features that distinguish melanoma from other solid tumors and contribute to the malignant phenotype of melanoma cells. For example, although melanoma cells are highly antigenic, they are extremely efficient at evading host immune response. Melanoma cells share numerous cell surface molecules with vascular cells, are highly angiogenic, are mesenchymal in nature, and possess a higher degree of 'stemness' than do other solid tumors. Finally, analysis of melanoma mutations has revealed that the gene expression profile of malignant melanoma is different from that of other cancers. Elucidating these molecular and genetic processes in highly metastatic melanoma can lead to the development of improved treatment and individualized therapy options.


Subject(s)
Antigens, Neoplasm , Gene Expression Regulation, Neoplastic , Melanoma , Neovascularization, Pathologic , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Gene Expression Profiling , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Organ Specificity
6.
PLoS One ; 8(5): e63221, 2013.
Article in English | MEDLINE | ID: mdl-23691000

ABSTRACT

BACKGROUND: The pharmaceutical and biotechnology industries depend on findings from academic investigators prior to initiating programs to develop new diagnostic and therapeutic agents to benefit cancer patients. The success of these programs depends on the validity of published findings. This validity, represented by the reproducibility of published findings, has come into question recently as investigators from companies have raised the issue of poor reproducibility of published results from academic laboratories. Furthermore, retraction rates in high impact journals are climbing. METHODS AND FINDINGS: To examine a microcosm of the academic experience with data reproducibility, we surveyed the faculty and trainees at MD Anderson Cancer Center using an anonymous computerized questionnaire; we sought to ascertain the frequency and potential causes of non-reproducible data. We found that ∼50% of respondents had experienced at least one episode of the inability to reproduce published data; many who pursued this issue with the original authors were never able to identify the reason for the lack of reproducibility; some were even met with a less than "collegial" interaction. CONCLUSIONS: These results suggest that the problem of data reproducibility is real. Biomedical science needs to establish processes to decrease the problem and adjudicate discrepancies in findings when they are discovered.


Subject(s)
Neoplasms/pathology , Translational Research, Biomedical , Data Collection , Humans , Neoplasms/diagnosis , Neoplasms/therapy , Reproducibility of Results
7.
Cancer Res ; 72(22): 5757-66, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22986745

ABSTRACT

Melanoma is the deadliest form of skin cancer in which patients with metastatic disease have a 5-year survival rate of less than 10%. Recently, the overexpression of a ß-galactoside binding protein, galectin-3 (LGALS3), has been correlated with metastatic melanoma in patients. We have previously shown that silencing galectin-3 in metastatic melanoma cells reduces tumor growth and metastasis. Gene expression profiling identified the protumorigenic gene autotaxin (ENPP2) to be downregulated after silencing galectin-3. Here we report that galectin-3 regulates autotaxin expression at the transcriptional level by modulating the expression of the transcription factor NFAT1 (NFATC2). Silencing galectin-3 reduced NFAT1 protein expression, which resulted in decreased autotaxin expression and activity. Reexpression of autotaxin in galectin-3 silenced melanoma cells rescues angiogenesis, tumor growth, and metastasis in vivo. Silencing NFAT1 expression in metastatic melanoma cells inhibited tumor growth and metastatic capabilities in vivo. Our data elucidate a previously unidentified mechanism by which galectin-3 regulates autotaxin and assign a novel role for NFAT1 during melanoma progression.


Subject(s)
Galectin 3/deficiency , Melanoma/pathology , NFATC Transcription Factors/biosynthesis , Phosphoric Diester Hydrolases/biosynthesis , Animals , Cell Line, Tumor , Female , Galectin 3/biosynthesis , Galectin 3/genetics , Gene Silencing , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Melanoma/blood supply , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , NFATC Transcription Factors/genetics , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphoric Diester Hydrolases/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transfection
8.
Cancer Metastasis Rev ; 31(3-4): 621-32, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22684365

ABSTRACT

The progression of melanoma toward the metastatic phenotype occurs in a defined stepwise manner. While many molecular changes take place early in melanoma development, progression toward the malignant phenotype, most notably during the transition from the radial growth phase (RGP) to the vertical growth phase (VGP) involves deregulated expression of several transcription factors. For example, the switch from RGP to VGP is associated with the loss of the transcription factor AP2α and gain of transcriptional activity of cAMP-responsive element binding protein. Together with the upregulation of microphthalmia-associated transcription factor, activating transcription factor 2, nuclear factor kappa B, and other transcription factors, these changes lead to dysregulated expression or function of important cellular adhesion molecules, matrix degrading enzymes, survival factors, as well as other factors leading to metastatic melanoma. Additionally, recent evidence suggests that microRNAs and RNA editing machinery influence the expression of transcription factors or are regulated themselves by transcription factors. Many of the downstream signaling molecules regulated by transcription factors, such as protease activated receptor-1, interleukin-8, and MCAM/MUC18 represent new treatment prospects.


Subject(s)
Melanoma/secondary , Transcription Factors/physiology , Activating Transcription Factor 1/physiology , Activating Transcription Factor 2/physiology , Animals , Cyclic AMP Response Element-Binding Protein/physiology , Gene Expression Regulation, Neoplastic , Humans , Melanoma/drug therapy , Microphthalmia-Associated Transcription Factor/physiology , NF-kappa B/physiology , RNA Editing , Transcription Factor AP-2/physiology
9.
Methods Mol Biol ; 814: 555-70, 2012.
Article in English | MEDLINE | ID: mdl-22144332

ABSTRACT

Astrocytes communicate with the vascular endothelium via direct cell-cell contacts as well as a variety of secreted growth factors and extracellular matrix (ECM) proteins. Integrins are heterodimeric cell surface receptors for ECM protein ligands, and many integrin subunits are expressed in astrocytes. Here, we will discuss gene deletion strategies in mice that have deciphered functions for specific integrins in astrocyte-endothelial cell adhesion and signaling. Specifically, we will detail how Cre-lox molecular genetic techniques have revealed important roles for integrin αvß8 in regulating cerebral blood vessel development and homeostasis. First, we will detail how to generate Cre-lox mutant mouse models that our group and others have used to study αvß8 integrin in embryonic astroglial progenitors and postnatal astrocytes. Second, we will discuss how viral-delivered Cre can be used to acutely delete integrin genes in astrocytes within defined anatomic regions of the brain. Third, detailed in vivo methods to verify Cre-mediated gene recombination in astrocytes will be presented. Lastly, we will present one experimental strategy to determine how integrin gene deletion affects astrocyte-endothelial cell coupling in the CNS. While this review focuses on the generation and characterization of mice lacking αvß8 integrin, these experimental strategies can be expanded to analyze other cell adhesion and signaling genes important for astroglial-mediated regulation of blood vessel development and homeostasis.


Subject(s)
Astrocytes/metabolism , Brain/cytology , Cell Adhesion/physiology , Cell Communication/physiology , Endothelial Cells/metabolism , Integrins/metabolism , Animals , Antibodies, Monoclonal , Astrocytes/physiology , Endothelial Cells/physiology , Extracellular Matrix Proteins/metabolism , Fluorescent Antibody Technique , Galactosides , Gene Deletion , Glial Fibrillary Acidic Protein , Indoles , Integrases , Integrins/genetics , Mice , Nerve Tissue Proteins/immunology
10.
Glia ; 59(11): 1579-87, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21674628

ABSTRACT

Neurogenesis in the post-natal brain occurs in two primary locations: the subgranular layer of the hippocampal dentate gyrus and the subventricular zone (SVZ) of the lateral ventricles. Following differentiation, neuroblasts within the SVZ migrate several millimeters to the olfactory bulbs (OBs) via a distinct anatomic route, or rostral migratory stream (RMS). The genes that govern neuroblast directional migration, and particularly those encoding cell adhesion and signaling factors, remain largely uncharacterized. Here, we report that the extracellular matrix adhesion receptor, ß8 integrin, is essential for proper neuroblast chain formation and directional navigation in the RMS. Primary neuroblasts isolated from the mouse brain express robust levels of ß8 integrin protein, and selective ablation of ß8 integrin gene expression in neuroblasts leads to aberrant chain migration and size-reduced OBs. These integrin-dependent defects can be recapitulated ex vivo using isolated neurospheres or SVZ explants. Collectively, these data identify essential cell-intrinsic functions for ß8 integrin in regulating neuroblast polarity and directional navigation in the mouse forebrain.


Subject(s)
Cell Movement/physiology , Integrin beta Chains/genetics , Integrin beta Chains/physiology , Neural Stem Cells/physiology , Neurogenesis/genetics , Neurogenesis/physiology , Neurons/physiology , Animals , Blotting, Western , Cell Movement/genetics , Cell Polarity/genetics , Cell Polarity/physiology , Cells, Cultured , Dentate Gyrus/cytology , Dentate Gyrus/physiology , Fluorescent Antibody Technique , Green Fluorescent Proteins , Immunohistochemistry , Immunoprecipitation , Lateral Ventricles/cytology , Lateral Ventricles/physiology , Mice , Mice, Knockout , Olfactory Bulb/cytology , Olfactory Bulb/growth & development
11.
J Cell Sci ; 122(Pt 11): 1842-51, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19461074

ABSTRACT

Central nervous system (CNS) neurovascular units are multicellular complexes consisting of neural cells, blood vessels and a milieu of extracellular matrix (ECM) proteins. ECM-mediated adhesion and signaling events within neurovascular units probably contribute to proper CNS development and physiology; however, the molecular mechanisms that control these events remain largely undetermined. Previous studies from our group and others showed that ablation of the ECM receptor, alphavbeta8 integrin, in neural progenitor cells (NPCs) of the embryonic mouse brain results in severe developmental neurovascular pathologies and premature death. Here, we have investigated the functions for this integrin in the adult brain by studying mice harboring a homozygous-null beta8 gene mutation generated on an outbred background that permits survival for several months. We show that adult beta8-/- mice display widespread defects in neurovascular unit homeostasis, including increased numbers of intracerebral blood vessels with pronounced perivascular astrogliosis. Furthermore, in neurogenic regions of the adult brain, where NPCs cluster around blood vessels in neurovascular niches, beta8 integrin is essential for normal control of NPC proliferation and survival. Analysis of NPCs cultured ex vivo reveals that the growth and survival defects correlate, in part, with diminished integrin-mediated activation of latent transforming growth factor beta1 (TGFbeta1), which is an ECM protein ligand for alphavbeta8 integrin. Collectively, these data identify essential functions for beta8 integrin in regulating neurovascular unit physiology in the post-natal mouse brain.


Subject(s)
Brain , Homeostasis , Integrins/metabolism , Neurogenesis/physiology , Animals , Blood-Brain Barrier/physiology , Brain/blood supply , Brain/cytology , Brain/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Extracellular Matrix/metabolism , Integrins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Olfactory Bulb/abnormalities , Olfactory Bulb/anatomy & histology , Phenotype , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism
12.
Leuk Res ; 32(5): 771-80, 2008 May.
Article in English | MEDLINE | ID: mdl-18031811

ABSTRACT

Epigenetic modifiers are currently in clinical use for various tumor types. Recently, numerous studies supporting the combination of histone deacetylase inhibitors (HDACi) and DNA methyltransferase inhibitors have emerged, encouraging early clinical trials of these agents together. Here we show that MS-275, an HDACi, and 5-azacytidine, a methyltransferase inhibitor, display synergistic cytotoxicity and apoptosis in AML and ALL cells. Intracellular production of reactive oxygen species (ROS), such as superoxide and hydrogen peroxide, is a novel marker for this synergism in ALL cells. These data suggest that assessment of oxidative stress can serve as a marker of the concerted action of MS-275 and 5-azacytidine.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Leukemia/drug therapy , Pyridines/pharmacology , Reactive Oxygen Species , Acetylation , Apoptosis/drug effects , Biomarkers , Caspase 3/physiology , Drug Synergism , Glutathione/metabolism , Histones/metabolism , Humans , Jurkat Cells , Leukemia/metabolism , Leukemia/pathology
SELECTION OF CITATIONS
SEARCH DETAIL