Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Mol Cancer Res ; 21(12): 1317-1328, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37606694

ABSTRACT

Although androgen deprivation treatment often effectively decreases prostate cancer, incurable metastatic castration-resistant prostate cancer (CRPC) eventually occurs. It is important to understand how CRPC metastasis progresses, which is not clearly defined. The loss of PTEN, a phosphatase to dephosphorylate phosphatidylinositol 3,4,5-trisphosphate in the PI3K pathway, occurs in up to 70% to 80% of CRPC. We generated a mouse androgen-independent prostate cancer cell line (PKO) from PTEN null and Hi-Myc transgenic mice in C57BL/6 background. We confirmed that this PKO cell line has an activated PI3K pathway and can metastasize into the femur and tibia of immunodeficient nude and immunocompetent C57BL/6 mice. In vitro, we found that androgen deprivation significantly enhanced PKO cell migration/invasion via the p110ß isoform-depended PAK1-MAPK activation. Inhibition of the p110ß-PAK1 axis significantly decreased prostate cancer cell migration/invasion. Of note, our analysis using clinical samples showed that PAK1 is more activated in CRPC than in advanced prostate cancer; high PAK1/phosphorylated-PAK1 levels are associated with decreased survival rates in patients with CRPC. All the information suggests that this cell line reflects the characteristics of CRPC cells and can be applied to dissect the mechanism of CRPC initiation and progression. This study also shows that PAK1 is a potential target for CRPC treatment. IMPLICATIONS: This study uses a newly generated PTEN null prostate cancer cell line to define a critical functional role of p110ß-PAK1 in CRPC migration/invasion. This study also shows that the p110ß-PAK1 axis can potentially be a therapeutic target in CRPC metastasis.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Animals , Humans , Male , Mice , Androgen Antagonists , Androgens/therapeutic use , Cell Line, Tumor , Mice, Inbred C57BL , Mice, Transgenic , p21-Activated Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Receptors, Androgen/metabolism
2.
Biochemistry ; 62(14): 2147-2160, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37403936

ABSTRACT

Werner syndrome protein (WRN) is a multifunctional enzyme with helicase, ATPase, and exonuclease activities that are necessary for numerous DNA-related transactions in the human cell. Recent studies identified WRN as a synthetic lethal target in cancers characterized by genomic microsatellite instability resulting from defects in DNA mismatch repair pathways. WRN's helicase activity is essential for the viability of these high microsatellite instability (MSI-H) cancers and thus presents a therapeutic opportunity. To this end, we developed a multiplexed high-throughput screening assay that monitors exonuclease, ATPase, and helicase activities of full-length WRN. This screening campaign led to the discovery of 2-sulfonyl/sulfonamide pyrimidine derivatives as novel covalent inhibitors of WRN helicase activity. The compounds are specific for WRN versus other human RecQ family members and show competitive behavior with ATP. Examination of these novel chemical probes established the sulfonamide NH group as a key driver of compound potency. One of the leading compounds, H3B-960, showed consistent activities in a range of assays (IC50 = 22 nM, KD = 40 nM, KI = 32 nM), and the most potent compound identified, H3B-968, has inhibitory activity IC50 ∼ 10 nM. These kinetic properties trend toward other known covalent druglike molecules. Our work provides a new avenue for screening WRN for inhibitors that may be adaptable to different therapeutic modalities such as targeted protein degradation, as well as a proof of concept for the inhibition of WRN helicase activity by covalent molecules.


Subject(s)
Neoplasms , Werner Syndrome , Humans , Exodeoxyribonucleases/genetics , RecQ Helicases/genetics , RecQ Helicases/metabolism , High-Throughput Screening Assays , Microsatellite Instability , Werner Syndrome Helicase/metabolism
3.
J Biol Chem ; 298(11): 102539, 2022 11.
Article in English | MEDLINE | ID: mdl-36179791

ABSTRACT

Recent studies have reported that the peroxisome proliferator-activated receptor gamma (PPARγ) pathway is activated in approximately 40% of patients with muscle-invasive bladder cancer. This led us to investigate pharmacological repression of PPARγ as a possible intervention strategy. Here, we characterize PPARγ antagonists and inverse agonists and find that the former behave as silent ligands, whereas inverse agonists (T0070907 and SR10221) repress downstream PPARγ target genes leading to growth inhibition in bladder cancer cell lines. To understand the mechanism, we determined the ternary crystal structure of PPARγ bound to T0070907 and the corepressor (co-R) peptide NCOR1. The structure shows that the AF-2 helix 12 (H12) rearranges to bind inside the ligand-binding domain, where it forms stabilizing interactions with the compound. This dramatic movement in H12 unveils a large interface for co-R binding. In contrast, the crystal structure of PPARγ bound to a SR10221 analog shows more subtle structural differences, where the compound binds and pushes H12 away from the ligand-binding domain to allow co-R binding. Interestingly, we found that both classes of compound promote recruitment of co-R proteins in biochemical assays but with distinct conformational changes in H12. We validate our structural models using both site-directed mutagenesis and chemical probes. Our findings offer new mechanistic insights into pharmacological modulation of PPARγ signaling.


Subject(s)
PPAR gamma , Urinary Bladder Neoplasms , Humans , PPAR gamma/metabolism , Ligands , Benzamides/pharmacology
4.
Mol Cancer Ther ; 21(6): 890-902, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35642432

ABSTRACT

Nearly 30% of patients with relapsed breast cancer present activating mutations in estrogen receptor alpha (ERα) that confer partial resistance to existing endocrine-based therapies. We previously reported the development of H3B-5942, a covalent ERα antagonist that engages cysteine-530 (C530) to achieve potency against both wild-type (ERαWT) and mutant ERα (ERαMUT). Anticipating that the emergence of C530 mutations could promote resistance to H3B-5942, we applied structure-based drug design to improve the potency of the core scaffold to further enhance the antagonistic activity in addition to covalent engagement. This effort led to the development of the clinical candidate H3B-6545, a covalent antagonist that is potent against both  ERαWT/MUT, and maintains potency even in the context of ERα C530 mutations. H3B-6545 demonstrates significant activity and superiority over standard-of-care fulvestrant across a panel of ERαWT and ERαMUT palbociclib sensitive and resistant models. In summary, the compelling preclinical activity of H3B-6545 supports its further development for the potential treatment of endocrine therapy-resistant ERα+ breast cancer harboring wild-type or mutant ESR1, as demonstrated by the ongoing clinical trials (NCT03250676, NCT04568902, NCT04288089). SUMMARY: H3B-6545 is an ERα covalent antagonist that exhibits encouraging preclinical activity against CDK4/6i naïve and resistant ERαWT and ERαMUT tumors.


Subject(s)
Breast Neoplasms , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Clinical Trials as Topic , Estrogen Receptor alpha/genetics , Female , Fulvestrant/therapeutic use , Humans , Indazoles , Neoplasm Recurrence, Local , Pyridines
5.
Mol Cancer Res ; 20(5): 673-685, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35105671

ABSTRACT

A common outcome of androgen deprivation in prostate cancer therapy is disease relapse and progression to castration-resistant prostate cancer (CRPC) via multiple mechanisms. To gain insight into the recent clinical findings that highlighted genomic alterations leading to hyperactivation of PI3K, we examined the roles of the commonly expressed p110 catalytic isoforms of PI3K in a murine model of Pten-null invasive CRPC. While blocking p110α had negligible effects in the development of Pten-null invasive CRPC, either genetic or pharmacologic perturbation of p110ß dramatically slowed CRPC initiation and progression. Once fully established, CRPC tumors became partially resistant to p110ß inhibition, indicating the acquisition of new dependencies. Driven by our genomic analyses highlighting potential roles for the p110ß/RAC/PAK1 and ß-catenin pathways in CRPC, we found that combining p110ß with RAC/PAK1 or tankyrase inhibitors significantly reduced the growth of murine and human CRPC organoids in vitro and in vivo. Because p110ß activity is dispensable for most physiologic processes, our studies support novel therapeutic strategies both for preventing disease progression into CRPC and for treating CRPC. IMPLICATIONS: This work establishes p110ß as a promising target for preventing the progression of primary PTEN-deficient prostate tumors to CRPC, and for treating established CRPC in combination with RAC/PAK1 or tankyrase inhibitors.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Tankyrases , Androgen Antagonists , Animals , Humans , Male , Mice , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases , Prostate , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics
7.
Cancer Res ; 79(8): 1740-1745, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30952631

ABSTRACT

The development of tamoxifen and subsequent estrogen receptor alpha (ERα) antagonists represents a tremendous therapeutic breakthrough in the treatment of breast cancer. Despite the ability of ERα antagonists to increase survival rates, resistance to these therapies is an all-too-common occurrence. The majority of resistant tumors, including those with hotspot mutations in the ligand-binding domain of ERα, remain dependent on ERα signaling, indicating that either a more potent or novel class of antagonist could have clinical benefit. With this thought in mind, we developed a novel ERα antagonist that exhibits enhanced potency due to its ability to covalently target a unique cysteine in ER. This review describes the design of this antagonist, H3B-5942, and discusses opportunities for future improvements, which could reduce the risk of escape mutations to this therapeutic modality.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Estrogen Receptor Antagonists/therapeutic use , Indazoles/therapeutic use , Receptors, Estrogen/antagonists & inhibitors , Animals , Female , Humans
8.
Cancer Chemother Pharmacol ; 83(1): 151-160, 2019 01.
Article in English | MEDLINE | ID: mdl-30386887

ABSTRACT

PURPOSE: H3B-6545, a novel selective estrogen receptor (ER)α covalent antagonist (SERCA) which inactivates both wild-type and mutant ERα, is in clinical development for the treatment of metastatic breast cancer. Preclinical studies were conducted to characterize the pharmacokinetics and metabolism of H3B-6545 in rat and monkeys. METHODS: The clearance and metabolic profiles of H3B-6545 were studied using rat, monkey and human hepatocytes, and reaction phenotyping was done using recombinant human cytochrome P450 enzymes. Blood stability, protein binding, and permeability were also determined in vitro. Pharmacokinetics of H3B-6545 was assessed after both intravenous and oral dosing. A nonclinical PBPK model was developed to assess in vitro-in vivo correlation of clearance. RESULTS: H3B-6545 had a terminal elimination half-life of 2.4 h in rats and 4.0 h in monkeys and showed low to moderate bioavailability, in line with the in vitro permeability assessment. Plasma protein binding was similar across species, at 99.5-99.8%. Nine metabolites of H3B-6545 were identified in hepatocyte incubations, none of which were unique to humans. Formation of glutathione-related conjugate of H3B-6545 was minimal in vitro. H3B-6545, a CYP3A substrate, is expected to be mostly cleared via hepatic phase 1 metabolism. Hepatocyte clearance values were used to adequately model the time-concentration profiles in rat and monkey. CONCLUSIONS: We report on the absorption and metabolic fate and disposition of H3B-6545 in rats and dogs and illustrate that in vitro-in vivo correlation of clearance is possible for targeted covalent inhibitors, provided reactivity is not a predominant mechanism of clearance.


Subject(s)
Cytochrome P-450 Enzyme System/drug effects , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor Antagonists/pharmacokinetics , Estrogen Receptor alpha/antagonists & inhibitors , Hepatocytes/metabolism , Indazoles/pharmacology , Indazoles/pharmacokinetics , Microsomes, Liver/metabolism , Pyridines/pharmacology , Pyridines/pharmacokinetics , Animals , Biological Availability , Cells, Cultured , Cytochrome P-450 Enzyme Inhibitors/pharmacokinetics , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Dogs , Drug Evaluation, Preclinical , Female , Hepatocytes/drug effects , Humans , In Vitro Techniques , Macaca fascicularis , Metabolic Clearance Rate , Microsomes, Liver/drug effects , Protein Binding , Rats, Sprague-Dawley , Species Specificity , Tissue Distribution
10.
Nat Commun ; 8(1): 103, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28740126

ABSTRACT

Muscle-invasive bladder cancer (MIBC) is an aggressive disease with limited therapeutic options. Although immunotherapies are approved for MIBC, the majority of patients fail to respond, suggesting existence of complementary immune evasion mechanisms. Here, we report that the PPARγ/RXRα pathway constitutes a tumor-intrinsic mechanism underlying immune evasion in MIBC. Recurrent mutations in RXRα at serine 427 (S427F/Y), through conformational activation of the PPARγ/RXRα heterodimer, and focal amplification/overexpression of PPARγ converge to modulate PPARγ/RXRα-dependent transcription programs. Immune cell-infiltration is controlled by activated PPARγ/RXRα that inhibits expression/secretion of inflammatory cytokines. Clinical data sets and an in vivo tumor model indicate that PPARγHigh/RXRαS427F/Y impairs CD8+ T-cell infiltration and confers partial resistance to immunotherapies. Knockdown of PPARγ or RXRα and pharmacological inhibition of PPARγ significantly increase cytokine expression suggesting therapeutic approaches to reviving immunosurveillance and sensitivity to immunotherapies. Our study reveals a class of tumor cell-intrinsic "immuno-oncogenes" that modulate the immune microenvironment of cancer.Muscle-invasive bladder cancer (MIBC) is a potentially lethal disease. Here the authors characterize diverse genetic alterations in MIBC that convergently lead to constitutive activation of PPARgamma/RXRalpha and result in immunosurveillance escape by inhibiting CD8+ T-cell recruitment.


Subject(s)
Immune Evasion/immunology , Monitoring, Immunologic , PPAR gamma/immunology , Retinoid X Receptor alpha/immunology , Urinary Bladder Neoplasms/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Gene Expression Profiling/methods , HCT116 Cells , Humans , Immunoblotting , Immunotherapy/methods , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Mice , Microscopy, Fluorescence , Mutation/immunology , Neoplasm Invasiveness , PPAR gamma/chemistry , PPAR gamma/genetics , Protein Multimerization/immunology , Retinoid X Receptor alpha/chemistry , Retinoid X Receptor alpha/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/therapy
11.
Nat Commun ; 8: 15522, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28541300

ABSTRACT

Pladienolide, herboxidiene and spliceostatin have been identified as splicing modulators that target SF3B1 in the SF3b subcomplex. Here we report that PHF5A, another component of this subcomplex, is also targeted by these compounds. Mutations in PHF5A-Y36, SF3B1-K1071, SF3B1-R1074 and SF3B1-V1078 confer resistance to these modulators, suggesting a common interaction site. RNA-seq analysis reveals that PHF5A-Y36C has minimal effect on basal splicing but inhibits the global action of splicing modulators. Moreover, PHF5A-Y36C alters splicing modulator-induced intron-retention/exon-skipping profile, which correlates with the differential GC content between adjacent introns and exons. We determine the crystal structure of human PHF5A demonstrating that Y36 is located on a highly conserved surface. Analysis of the cryo-EM spliceosome Bact complex shows that the resistance mutations cluster in a pocket surrounding the branch point adenosine, suggesting a competitive mode of action. Collectively, we propose that PHF5A-SF3B1 forms a central node for binding to these splicing modulators.


Subject(s)
Adenosine/chemistry , Alternative Splicing , Carrier Proteins/chemistry , Phosphoproteins/chemistry , RNA Splicing Factors/chemistry , Cell Proliferation , Cell Survival , Cryoelectron Microscopy , Crystallography, X-Ray , Epoxy Compounds/chemistry , Exons , Fatty Alcohols/chemistry , HCT116 Cells , Humans , Introns , Macrolides/chemistry , Mass Spectrometry , Mutagenesis, Site-Directed , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Phosphoproteins/metabolism , Protein Binding , Protein Conformation , Pyrans/chemistry , RNA Interference , RNA Splicing Factors/metabolism , RNA-Binding Proteins , Recombinant Proteins/chemistry , Sequence Analysis, RNA , Spiro Compounds/chemistry , Spliceosomes/metabolism , Trans-Activators
12.
J Vis Exp ; (106): e52879, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26710000

ABSTRACT

Although targeted therapies are initially effective, resistance inevitably emerges. Several methods, such as genetic analysis of resistant clinical specimens, have been applied to uncover these resistance mechanisms to facilitate follow-up care. Although these approaches have led to clinically relevant discoveries, difficulties in attaining the relevant patient material or in deconvoluting the genomic data collected from these specimens have severely hampered the path towards a cure. To this end, we here describe a tool for expeditious discovery that may guide improvement in first-line therapies and alternative clinical management strategies. By coupling preclinical in vitro or in vivo drug selection with next-generation sequencing, it is possible to identify genomic structural variations and/or gene expression alterations that may serve as functional drivers of resistance. This approach facilitates the spontaneous emergence of alterations, enhancing the probability that these mechanisms may be observed in the patients. In this protocol we provide guidelines to maximize the potential for uncovering single nucleotide variants that drive resistance using adherent lines.


Subject(s)
Drug Resistance/genetics , High-Throughput Nucleotide Sequencing/methods , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , HCT116 Cells , Humans , In Vitro Techniques , Molecular Targeted Therapy
13.
Cancer Discov ; 3(9): 1030-43, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23842682

ABSTRACT

UNLABELLED: Castration-resistant prostate cancer (CRPC) is the most aggressive, incurable form of prostate cancer. MDV3100 (enzalutamide), an antagonist of the androgen receptor (AR), was approved for clinical use in men with metastatic CRPC. Although this compound showed clinical efficacy, many initial responders later developed resistance. To uncover relevant resistant mechanisms, we developed a model of spontaneous resistance to MDV3100 in LNCaP prostate cancer cells. Detailed characterization revealed that emergence of an F876L mutation in AR correlated with blunted AR response to MDV3100 and sustained proliferation during treatment. Functional studies confirmed that AR(F876L) confers an antagonist-to-agonist switch that drives phenotypic resistance. Finally, treatment with distinct antiandrogens or cyclin-dependent kinase (CDK)4/6 inhibitors effectively antagonized AR(F876L) function. Together, these findings suggest that emergence of F876L may (i) serve as a novel biomarker for prediction of drug sensitivity, (ii) predict a "withdrawal" response to MDV3100, and (iii) be suitably targeted with other antiandrogens or CDK4/6 inhibitors. SIGNIFICANCE: We uncovered an F876L agonist-switch mutation in AR that confers genetic and phenotypic resistance to the antiandrogen drug MDV3100. On the basis of this fi nding, we propose new therapeutic strategies to treat patients with prostate cancer presenting with this AR mutation.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/genetics , Androgen Antagonists/pharmacology , Base Sequence , Benzamides , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Humans , Male , Mutation , Nitriles , Phenylthiohydantoin/pharmacology , Sequence Analysis, DNA
14.
J Biol Chem ; 287(50): 42180-94, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23038259

ABSTRACT

Mutations in the genes encoding isocitrate dehydrogenase 1 and 2 (IDH1/2) occur in a variety of tumor types, resulting in production of the proposed oncometabolite, 2-hydroxyglutarate (2-HG). How mutant IDH and 2-HG alter signaling pathways to promote cancer, however, remains unclear. Additionally, there exist relatively few cell lines with IDH mutations. To examine the effect of endogenous IDH mutations and 2-HG, we created a panel of isogenic epithelial cell lines with either wild-type IDH1/2 or clinically relevant IDH1/2 mutations. Differences were noted in the ability of IDH mutations to cause robust 2-HG accumulation. IDH1/2 mutants that produce high levels of 2-HG cause an epithelial-mesenchymal transition (EMT)-like phenotype, characterized by changes in EMT-related gene expression and cellular morphology. 2-HG is sufficient to recapitulate aspects of this phenotype in the absence of an IDH mutation. In the cells types examined, mutant IDH-induced EMT is dependent on up-regulation of the transcription factor ZEB1 and down-regulation of the miR-200 family of microRNAs. Furthermore, sustained knockdown of IDH1 in IDH1 R132H mutant cells is sufficient to reverse many characteristics of EMT, demonstrating that continued expression of mutant IDH is required to maintain this phenotype. These results suggest mutant IDH proteins can reversibly deregulate discrete signaling pathways that contribute to tumorigenesis.


Subject(s)
Epithelial-Mesenchymal Transition , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/metabolism , Isocitrate Dehydrogenase/biosynthesis , MicroRNAs/biosynthesis , Mutation, Missense , Neoplasm Proteins/metabolism , Neoplasms/metabolism , RNA, Neoplasm/biosynthesis , Transcription Factors/metabolism , Amino Acid Substitution , Cell Line, Tumor , Glutarates/metabolism , Homeodomain Proteins/genetics , Humans , Isocitrate Dehydrogenase/genetics , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , RNA, Neoplasm/genetics , Transcription Factors/genetics , Up-Regulation/genetics , Zinc Finger E-box-Binding Homeobox 1
15.
Nat Med ; 17(9): 1101-8, 2011 Aug 07.
Article in English | MEDLINE | ID: mdl-21822286

ABSTRACT

Although the role of miR-200s in regulating E-cadherin expression and epithelial-to-mesenchymal transition is well established, their influence on metastatic colonization remains controversial. Here we have used clinical and experimental models of breast cancer metastasis to discover a pro-metastatic role of miR-200s that goes beyond their regulation of E-cadherin and epithelial phenotype. Overexpression of miR-200s is associated with increased risk of metastasis in breast cancer and promotes metastatic colonization in mouse models, phenotypes that cannot be recapitulated by E-cadherin expression alone. Genomic and proteomic analyses revealed global shifts in gene expression upon miR-200 overexpression toward that of highly metastatic cells. miR-200s promote metastatic colonization partly through direct targeting of Sec23a, which mediates secretion of metastasis-suppressive proteins, including Igfbp4 and Tinagl1, as validated by functional and clinical correlation studies. Overall, these findings suggest a pleiotropic role of miR-200s in promoting metastatic colonization by influencing E-cadherin-dependent epithelial traits and Sec23a-mediated tumor cell secretome.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , MicroRNAs/metabolism , Neoplasm Metastasis/physiopathology , Vesicular Transport Proteins/metabolism , Animals , Cadherins/metabolism , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , Mass Spectrometry , Mice , Mice, Inbred BALB C , Microarray Analysis , Statistics, Nonparametric
16.
J Transl Med ; 9: 30, 2011 Mar 22.
Article in English | MEDLINE | ID: mdl-21426561

ABSTRACT

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is often diagnosed at later stages until they are incurable. MicroRNA (miR) is a small, non-coding RNA that negatively regulates gene expression mainly via translational repression. Accumulating evidence indicates that deregulation of miR is associated with human malignancies including ESCC. The aim of this study was to identify miR that could be specifically expressed and exert distinct biological actions in ESCC. METHODS: Total RNA was extracted from ESCC cell lines, OE21 and TE10, and a non-malignant human esophageal squamous cell line, Het-1A, and subjected to microarray analysis. Expression levels of miR that showed significant differences between the 2 ESCC and Het-1A cells based on the comprehensive analysis were analyzed by the quantitative reverse transcriptase (RT)-PCR method. Then, functional analyses, including cellular proliferation, apoptosis and Matrigel invasion and the wound healing assay, for the specific miR were conducted. Using ESCC tumor samples and paired surrounding non-cancerous tissue obtained endoscopically, the association with histopathological differentiation was examined with quantitative RT-PCR. RESULTS: Based on the miR microarray analysis, there were 14 miRs that showed significant differences (more than 2-fold) in expression between the 2 ESCC cells and non-malignant Het-1A. Among the significantly altered miRs, miR-205 expression levels were exclusively higher in 5 ESCC cell lines examined than any other types of malignant cell lines and Het-1A. Thus, miR-205 could be a specific miR in ESCC. Modulation of miR-205 expression by transfection with its precursor or anti-miR-205 inhibitor did not affect ESCC cell proliferation and apoptosis, but miR-205 was found to be involved in cell invasion and migration. Western blot revealed that knockdown of miR-205 expression in ESCC cells substantially enhanced expression of zinc finger E-box binding homeobox 2, accompanied by reduction of E-cadherin, a regulator of epithelial mesenchymal transition. The miR-205 expression levels were not associated with histological differentiation of human ESCC. CONCLUSIONS: These results imply that miR-205 is an ESCC-specific miR that exerts tumor-suppressive activities with EMT inhibition by targeting ZEB2.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cell Movement/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , MicroRNAs/metabolism , Repressor Proteins/genetics , Apoptosis/genetics , Carcinoma, Squamous Cell/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Homeodomain Proteins/metabolism , Humans , MicroRNAs/genetics , Neoplasm Invasiveness , Phenotype , Repressor Proteins/metabolism , Up-Regulation/genetics , Zinc Finger E-box Binding Homeobox 2
17.
Eur J Cancer ; 46(7): 1232-40, 2010 May.
Article in English | MEDLINE | ID: mdl-20307969

ABSTRACT

Transforming growth factor (TGF)-beta signalling plays a dichotomous role in tumour progression, acting as a tumour suppressor early and as a pro-metastatic pathway in late-stages. There is accumulating evidence that advanced-stage tumours produce excessive levels of TGF-beta, which acts to promote tumour growth, invasion and colonisation of secondary organs. In light of the pro-metastasis function, many strategies are currently being explored to antagonise the TGF-beta pathway as a treatment for metastatic cancers. Strategies such as using large molecule ligand traps, reducing the translational efficiency of TGF-beta ligands using antisense technology, and antagonising TGF-beta receptor I/II kinase function using small molecule inhibitors are the most prominent methods being explored today. Administration of anti-TGF-beta therapies alone, or in combination with immunosuppressive or cytotoxic therapies, has yielded promising results in the preclinical and clinical settings. Despite these successes, the temporal- and context-dependent roles of TGF-beta signalling in cancer has made it challenging to define patient subgroups that are most likely to respond, and the therapeutic regimens that will be most effective in the clinic. Novel mouse models and diagnostic tools are being developed today to circumvent these issues, which may potentially expedite anti-TGF-beta drug development and clinical application.


Subject(s)
Neoplasm Metastasis/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Antisense Elements (Genetics)/therapeutic use , Cell Transformation, Neoplastic/metabolism , Combined Modality Therapy , Disease Progression , Humans , Mice , Neoplasm Metastasis/physiopathology , Neoplasm Proteins/immunology , Neoplasm Proteins/physiology , Signal Transduction/drug effects , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/physiology
18.
Nat Med ; 15(8): 960-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19597504

ABSTRACT

Although the transforming growth factor-beta (TGF-beta) pathway has been implicated in breast cancer metastasis, its in vivo dynamics and temporal-spatial involvement in organ-specific metastasis have not been investigated. Here we engineered a xenograft model system with a conditional control of the TGF-beta-SMAD signaling pathway and a dual-luciferase reporter system for tracing both metastatic burden and TGF-beta signaling activity in vivo. Strong TGF-beta signaling in osteolytic bone lesions is suppressed directly by genetic and pharmacological disruption of the TGF-beta-SMAD pathway and indirectly by inhibition of osteoclast function with bisphosphonates. Notably, disruption of TGF-beta signaling early in metastasis can substantially reduce metastasis burden but becomes less effective when bone lesions are well established. Our in vivo system for real-time manipulation and detection of TGF-beta signaling provides a proof of principle for using similar strategies to analyze the in vivo dynamics of other metastasis-associated signaling pathways and will expedite the development and characterization of therapeutic agents.


Subject(s)
Bone Neoplasms/diagnosis , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Diagnostic Imaging/methods , Transforming Growth Factor beta/pharmacokinetics , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/therapy , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Female , Genetic Vectors , Humans , Mice , Mice, Nude , Models, Biological , Signal Transduction/drug effects , Smad4 Protein/genetics , Smad4 Protein/metabolism , Treatment Outcome , Tumor Cells, Cultured , Validation Studies as Topic , Xenograft Model Antitumor Assays
19.
J Biol Chem ; 283(22): 14910-4, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18411277

ABSTRACT

MicroRNAs are small non-coding RNA molecules that can regulate gene expression by interacting with multiple mRNAs and inducing either translation suppression or degradation of mRNA. Recently, several miRNAs were identified as either promoters or suppressors of metastasis. However, it is unclear in which step(s) of the multistep metastatic cascade these miRNAs play a defined functional role. To study the functional importance of miRNAs in epithelial-mesenchymal transition (EMT), a process thought to initiate metastasis by enhancing the motility of tumor cells, we used a well established in vitro EMT assay: transforming growth factor-beta-induced EMT in NMuMG murine mammary epithelial cells. We found that members of the miR-200 family, organized as two clusters in the genome, were repressed during EMT. Overexpression of each miRNA individually or as clusters in NMuMG cells hindered EMT by enhancing E-cadherin expression through direct targeting of ZEB1 and ZEB2, which encode transcriptional repressors of E-cadherin. In the 4TO7 mouse carcinoma cell line, which expresses low levels of endogenous E-cadherin and displays a mesenchymal phenotype, ectopic expression of the miR-200 family miRNAs significantly increased E-cadherin expression and altered cell morphology to an epithelial phenotype. Furthermore, ectopic expression of each miR-200 miRNA cluster significantly reduced the in vitro motility of 4TO7 cells in migration assays. These results suggested that loss of expression of the miR-200 family members may play a critical role in the repression of E-cadherin by ZEB1 and ZEB2 during EMT, thereby enhancing migration and invasion during cancer progression.


Subject(s)
Cadherins/biosynthesis , Cell Movement , Homeodomain Proteins/biosynthesis , Kruppel-Like Transcription Factors/biosynthesis , MicroRNAs/biosynthesis , Neoplasms/metabolism , Repressor Proteins/biosynthesis , Transcription Factors/biosynthesis , Animals , Cadherins/genetics , Cell Movement/drug effects , Cell Movement/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Genome, Human/genetics , HeLa Cells , Homeodomain Proteins/genetics , Humans , Kruppel-Like Transcription Factors/genetics , Mice , MicroRNAs/genetics , Multigene Family/genetics , Neoplasm Metastasis , Neoplasms/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/pharmacology , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
20.
J Biol Chem ; 283(20): 14109-19, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18347012

ABSTRACT

Microtubule spindle assembly in mitosis is stimulated by Ran.GTP, which is generated along condensed chromosomes by the guanine nucleotide exchange factor (GEF) RCC1. This relationship suggests that similar activities might modulate other microtubule structures. Interphase microtubules usually extend from the centrosome, although noncentrosomal microtubules function in some differentiated cells, including megakaryocytes. In these cells, platelet biogenesis requires massive mobilization of microtubules in the cell periphery, where they form proplatelets, the immediate precursors of platelets, in the apparent absence of centrioles. Here we identify a cytoplasmic Ran-binding protein, RanBP10, as a factor that binds beta-tubulin and associates with megakaryocyte microtubules. Unexpectedly, RanBP10 harbors GEF activity toward Ran. A point mutation in the candidate GEF domain abolishes exchange activity, and our results implicate RanBP10 as a localized cytoplasmic Ran-GEF. RNA interference-mediated loss of RanBP10 in cultured megakaryocytes disrupts microtubule organization. These results lead us to propose that spatiotemporally restricted generation of cytoplasmic Ran.GTP may influence organization of the specialized microtubules required in thrombopoiesis and that RanBP10 might serve as a molecular link between Ran and noncentrosomal microtubules.


Subject(s)
Cytoplasm/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Microtubule-Associated Proteins/physiology , Microtubules/metabolism , ran GTP-Binding Protein/metabolism , Animals , Blood Platelets/metabolism , Cell Differentiation , Centrosome/chemistry , Centrosome/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/physiology , Humans , Megakaryocytes/metabolism , Mice , Microtubule-Associated Proteins/genetics , Models, Biological , RNA Interference , Thrombopoiesis , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...