Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
1.
Leukemia ; 32(3): 774-787, 2018 03.
Article in English | MEDLINE | ID: mdl-28804127

ABSTRACT

T-cell prolymphocytic leukemia (T-PLL) is a rare and aggressive neoplasm of mature T-cells with an urgent need for rationally designed therapies to address its notoriously chemo-refractory behavior. The median survival of T-PLL patients is <2 years and clinical trials are difficult to execute. Here we systematically explored the diversity of drug responses in T-PLL patient samples using an ex vivo drug sensitivity and resistance testing platform and correlated the findings with somatic mutations and gene expression profiles. Intriguingly, all T-PLL samples were sensitive to the cyclin-dependent kinase inhibitor SNS-032, which overcame stromal-cell-mediated protection and elicited robust p53-activation and apoptosis. Across all patients, the most effective classes of compounds were histone deacetylase, phosphoinositide-3 kinase/AKT/mammalian target of rapamycin, heat-shock protein 90 and BH3-family protein inhibitors as well as p53 activators, indicating previously unexplored, novel targeted approaches for treating T-PLL. Although Janus-activated kinase-signal transducer and activator of transcription factor (JAK-STAT) pathway mutations were common in T-PLL (71% of patients), JAK-STAT inhibitor responses were not directly linked to those or other T-PLL-specific lesions. Overall, we found that genetic markers do not readily translate into novel effective therapeutic vulnerabilities. In conclusion, novel classes of compounds with high efficacy in T-PLL were discovered with the comprehensive ex vivo drug screening platform warranting further studies of synergisms and clinical testing.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Leukemia, Prolymphocytic, T-Cell/genetics , Mutation , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Cell Cycle/genetics , Cell Line, Tumor , Chromosome Aberrations , Female , Gene Expression , Gene Expression Profiling , Humans , Janus Kinases/metabolism , Leukemia, Prolymphocytic, T-Cell/drug therapy , Leukemia, Prolymphocytic, T-Cell/metabolism , Male , Middle Aged , Molecular Targeted Therapy , Oxazoles/pharmacology , Phenotype , Protein Kinase Inhibitors/pharmacology , STAT Transcription Factors/metabolism , Thiazoles/pharmacology
2.
Leukemia ; 31(1): 51-57, 2017 01.
Article in English | MEDLINE | ID: mdl-27461063

ABSTRACT

TCF3-PBX1 (E2A-PBX1) is a recurrent gene fusion in B-cell precursor acute lymphoblastic leukemia (BCP-ALL), which is caused by the translocation t(1;19)(q23;p13). TCF3-PBX1 BCP-ALL patients typically benefit from chemotherapy; however, many relapse and subsequently develop resistant disease with few effective treatment options. Mechanisms driving disease progression and therapy resistance have not been studied in TCF3-PBX1 BCP-ALL. Here, we aimed to identify novel treatment options for TCF3-PBX1 BCP-ALL by profiling leukemia cells from a relapsed patient, and determine molecular mechanisms underlying disease pathogenesis and progression. By drug-sensitivity testing of leukemic blasts from the index patient, control samples and TCF3-PBX1 positive and negative BCP-ALL cell lines, we identified the phosphatidylinositide 3-kinase delta (p110δ) inhibitor idelalisib as an effective treatment for TCF3-PBX1 BCP-ALL. This was further supported by evidence showing TCF3-PBX1 directly regulates expression of PIK3CD, the gene encoding p110δ. Other somatic mutations to TP53 and MTOR, as well as aberrant expression of CXCR4, may influence additional drug sensitivities specific to the index patient and accompanied progression of the disease. Our results suggest that idelalisib is a promising treatment option for patients with TCF3-PBX1 BCP-ALL, whereas other drugs could be useful depending on the genetic context of individual patients.


Subject(s)
Oncogene Proteins, Fusion , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Purines/pharmacology , Quinazolinones/pharmacology , Adult , Cell Line, Tumor , Class Ia Phosphatidylinositol 3-Kinase/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Male , Oncogene Proteins, Fusion/physiology , Phosphoinositide-3 Kinase Inhibitors , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Purines/therapeutic use , Quinazolinones/therapeutic use , Recurrence
3.
Leukemia ; 31(2): 301-309, 2017 02.
Article in English | MEDLINE | ID: mdl-27499136

ABSTRACT

Inhibitors of B-cell lymphoma-2 (BCL-2) such as venetoclax (ABT-199) and navitoclax (ABT-263) are clinically explored in several cancer types, including acute myeloid leukemia (AML), to selectively induce apoptosis in cancer cells. To identify robust biomarkers for BCL-2 inhibitor sensitivity, we evaluated the ex vivo sensitivity of fresh leukemic cells from 73 diagnosed and relapsed/refractory AML patients, and then comprehensively assessed whether the responses correlated to specific mutations or gene expression signatures. Compared with samples from healthy donor controls (nonsensitive) and chronic lymphocytic leukemia (CLL) patients (highly sensitive), AML samples exhibited variable responses to BCL-2 inhibition. Strongest CLL-like responses were observed in 15% of the AML patient samples, whereas 32% were resistant, and the remaining exhibited intermediate responses to venetoclax. BCL-2 inhibitor sensitivity was associated with genetic aberrations in chromatin modifiers, WT1 and IDH1/IDH2. A striking selective overexpression of specific HOXA and HOXB gene transcripts were detected in highly BCL-2 inhibitor sensitive samples. Ex vivo responses to venetoclax showed significant inverse correlation to ß2-microglobulin expression and to a lesser degree to BCL-XL and BAX expression. As new therapy options for AML are urgently needed, the specific HOX gene expression pattern can potentially be used as a biomarker to identify venetoclax-sensitive AML patients for clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Leukemic , Genes, Homeobox , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Aniline Compounds/pharmacology , Antineoplastic Agents/therapeutic use , Biopsy , Bone Marrow/pathology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Case-Control Studies , Cell Line, Tumor , Cluster Analysis , Drug Resistance, Neoplasm/genetics , Exome , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Multigene Family , Mutation , Sulfonamides/pharmacology , WT1 Proteins/genetics , beta 2-Microglobulin/genetics
4.
Leukemia ; 31(5): 1048-1058, 2017 05.
Article in English | MEDLINE | ID: mdl-27795554

ABSTRACT

In our individualized systems medicine program, personalized treatment options are identified and administered to chemorefractory acute myeloid leukemia (AML) patients based on exome sequencing and ex vivo drug sensitivity and resistance testing data. Here, we analyzed how clonal heterogeneity affects the responses of 13 AML patients to chemotherapy or targeted treatments using ultra-deep (average 68 000 × coverage) amplicon resequencing. Using amplicon resequencing, we identified 16 variants from 4 patients (frequency 0.54-2%) that were not detected previously by exome sequencing. A correlation-based method was developed to detect mutation-specific responses in serial samples across multiple time points. Significant subclone-specific responses were observed for both chemotherapy and targeted therapy. We detected subclonal responses in patients where clinical European LeukemiaNet (ELN) criteria showed no response. Subclonal responses also helped to identify putative mechanisms underlying drug sensitivities, such as sensitivity to azacitidine in DNMT3A mutated cell clones and resistance to cytarabine in a subclone with loss of NF1 gene. In summary, ultra-deep amplicon resequencing method enables sensitive quantification of subclonal variants and their responses to therapies. This approach provides new opportunities for designing combinatorial therapies blocking multiple subclones as well as for real-time assessment of such treatments.


Subject(s)
Clone Cells/drug effects , Leukemia, Myeloid, Acute/drug therapy , Antineoplastic Agents/pharmacology , Base Sequence , Drug Monitoring , Genetic Variation , Humans , Leukemia, Myeloid, Acute/genetics , Molecular Targeted Therapy , Precision Medicine
5.
Eur J Pharm Sci ; 98: 30-39, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-27771514

ABSTRACT

Urinary extracellular vesicles (EVs) are a promising source of biomarkers, which can be obtained in a non-invasive manner. However, the yield of EVs from urine samples may be insufficient for various analyses due to the entrapment of EVs by the Tamm-Horsfall protein (THP) meshwork. Here, we developed a simple dilution protocol to increase the urinary EV yield by disrupting the interaction between THP filaments and EVs with the help of alkaline pH and lowered ionic concentration. The integrity of the EVs and THP was assessed by electron microscopy. The effect of the protocol on the EV yield was quantified against an undiluted control by western blotting of four EV markers, nanoparticle tracking analysis and measuring of the RNA/miRNA concentration of the EV samples. The average EV yield from the dilution protocol was 2-7 fold the yield from the undiluted control i.e. increased by 130-624% as measured by western blotting and NTA. The yield increased most from samples with a high THP to EV ratio. The morphology and size range of the EVs were unaltered by the protocol. However, RNA/miRNA yields were the same as from the undiluted control and THP filaments could still be detected in EV samples. The dilution protocol, that we named KeepEX, provides a simple and efficient way to prevent loss of EVs thus increasing their yield from urine. Since KeepEX does not require individual adjustment of sample pH nor extra centrifugation steps, it could be used on its own or in combination with other EV purification protocols to improve EV isolation particularly from small urine volumes.


Subject(s)
Extracellular Vesicles , Urine/cytology , Centrifugation , Extracellular Vesicles/ultrastructure , Humans , Hydrogen-Ion Concentration , Microscopy, Electron, Transmission , Proteins/analysis , Tromethamine/chemistry
6.
Leukemia ; 31(5): 1187-1195, 2017 05.
Article in English | MEDLINE | ID: mdl-27833094

ABSTRACT

We sought to identify drugs that could counteract cytarabine resistance in acute myeloid leukemia (AML) by generating eight resistant variants from MOLM-13 and SHI-1 AML cell lines by long-term drug treatment. These cells were compared with 66 ex vivo chemorefractory samples from cytarabine-treated AML patients. The models and patient cells were subjected to genomic and transcriptomic profiling and high-throughput testing with 250 emerging and clinical oncology compounds. Genomic profiling uncovered deletion of the deoxycytidine kinase (DCK) gene in both MOLM-13- and SHI-1-derived cytarabine-resistant variants and in an AML patient sample. Cytarabine-resistant SHI-1 variants and a subset of chemorefractory AML patient samples showed increased sensitivity to glucocorticoids that are often used in treatment of lymphoid leukemia but not AML. Paired samples taken from AML patients before treatment and at relapse also showed acquisition of glucocorticoid sensitivity. Enhanced glucocorticoid sensitivity was only seen in AML patient samples that were negative for the FLT3 mutation (P=0.0006). Our study shows that development of cytarabine resistance is associated with increased sensitivity to glucocorticoids in a subset of AML, suggesting a new therapeutic strategy that should be explored in a clinical trial of chemorefractory AML patients carrying wild-type FLT3.


Subject(s)
Cytarabine/pharmacology , Drug Resistance, Neoplasm , Glucocorticoids/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Adult , Cytarabine/therapeutic use , Gene Expression Profiling , Humans , Tumor Cells, Cultured , Young Adult , fms-Like Tyrosine Kinase 3/genetics
7.
Blood Cancer J ; 5: e309, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25933373

ABSTRACT

Chronic myeloid leukemia in blast crisis (CML BC) remains a challenging disease to treat despite the introduction and advances in tyrosine kinase inhibitor (TKI) therapy. In this study we set out to identify novel candidate drugs for CML BC by using an unbiased high-throughput drug testing platform. We used three CML cell lines representing different types of CML blast phases (K562, EM-2 and MOLM-1) and primary leukemic cells from three CML BC patients. Profiling of drug responses was performed with a drug sensitivity and resistance testing platform comprising 295 anticancer agents. Overall, drug sensitivity scores and the drug response profiles of cell line and primary cell samples correlated well and were distinct from other types of leukemia samples. The cell lines were highly sensitive to TKIs and the clinically TKI-resistant patient samples were also resistant ex vivo. Comparison of cell line and patient sample data identified new candidate drugs for CML BC, such as vascular endothelial growth factor receptor and nicotinamide phosphoribosyltransferase inhibitors. Our results indicate that these drugs in particular warrant further evaluation by analyzing a larger set of primary patient samples. The results also pave way for designing rational combination therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Blast Crisis/drug therapy , Cell Survival/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Adult , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , High-Throughput Screening Assays , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
8.
Cell Death Dis ; 5: e1043, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24481457

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/TNFSF10/Apo2L) holds promise for cancer therapy as it induces apoptosis in a large variety of cancer cells while exerting negligible toxicity in normal ones. However, TRAIL can also induce proliferative and migratory signaling in cancer cells resistant to apoptosis induced by this cytokine. In that regard, the molecular mechanisms underlying the tumor selectivity of TRAIL and those balancing apoptosis versus survival remain largely elusive. We show here that high mRNA levels of PLAU, which encodes urokinase plasminogen activator (uPA), are characteristic of cancer cells with functional TRAIL signaling. Notably, decreasing uPA levels sensitized cancer cells to TRAIL, leading to markedly increased apoptosis. Mechanistic analyses revealed three molecular events taking place in uPA-depleted cells: reduced basal ERK1/2 prosurvival signaling, decreased preligand decoy receptor 2 (DcR2)-death receptor 5 (DR5) interaction and attenuated recruitment of DcR2 to the death-inducing signaling complex upon TRAIL challenge. These phenomena were accompanied by increased FADD and procaspase-8 recruitment and processing, thus guiding cells toward a caspase-dependent cell death that is largely independent of the intrinsic apoptosis pathway. Collectively, our results unveil PLAU mRNA levels as marker for the identification of TRAIL-responsive tumor cells and highlight a key role of uPA signaling in 'apoptosis versus survival' decision-making processes upon TRAIL challenge.


Subject(s)
Neoplasms/enzymology , Neoplasms/physiopathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Urokinase-Type Plasminogen Activator/genetics , Apoptosis , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival , Humans , Neoplasms/genetics , Protein Binding , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction , Urokinase-Type Plasminogen Activator/metabolism
10.
Oncogene ; 33(27): 3538-49, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24013224

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising agent in selectively killing tumor cells. However, TRAIL monotherapy has not been successful as many cancer cells are resistant to TRAIL. Chemotherapeutic agents, such as doxorubicin have been shown to act synergistically with TRAIL, but the exact mechanisms of actions are poorly understood. In this study, we performed high-throughput small interfering RNA screening and genome-wide gene expression profiling on doxorubicin-treated U1690 cells to explore novel mechanisms underlying doxorubicin-TRAIL synergy. The screening and expression profiling results were integrated and dihydroorotate dehydrogenase (DHODH) was identified as a potential candidate. DHODH is the rate-limiting enzyme in the pyrimidine synthesis pathway, and its expression was downregulated by doxorubicin. We demonstrated that silencing of DHODH or inhibition of DHODH activity by brequinar dramatically increased the sensitivity of U1690 cells to TRAIL-induced apoptosis both in 2D and 3D cultures, and was accompanied by downregulation of c-FLIPL as well as by mitochondrial depolarization. In addition, uridine, an end product of the pyrimidine synthesis pathway was able to rescue the sensitization effects initiated by both brequinar and doxorubicin. Furthermore, several other cancer cell lines, LNCaP, MCF-7 and HT-29 were also shown to be sensitized to TRAIL by brequinar. Taken together, our findings have identified a novel protein target and its inhibitor, brequinar, as a potential agent in TRAIL-based combinatorial cancer therapy and highlighted for the first time the importance of mitochondrial DHODH enzyme and pyrimidine pathway in mediating TRAIL sensitization in cancer cells.


Subject(s)
Biphenyl Compounds/pharmacology , Doxorubicin/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Pyrimidines/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Computational Biology , Dihydroorotate Dehydrogenase , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Gene Expression Profiling , Gene Silencing , Humans , Oxidoreductases Acting on CH-CH Group Donors/deficiency , Oxidoreductases Acting on CH-CH Group Donors/genetics , RNA, Small Interfering/genetics , Uridine/pharmacology
11.
Blood Cancer J ; 3: e168, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24317090

ABSTRACT

T-cell large granular lymphocytic (T-LGL) leukemia is a clonal disease characterized by the expansion of mature CD3+CD8+ cytotoxic T cells. It is often associated with autoimmune disorders and immune-mediated cytopenias. Our recent findings suggest that up to 40% of T-LGL patients harbor mutations in the STAT3 gene, whereas STAT5 mutations are present in 2% of patients. In order to identify putative disease-causing genetic alterations in the remaining T-LGL patients, we performed exome sequencing from three STAT mutation-negative patients and validated the findings in 113 large granular lymphocytic (LGL) leukemia patients. On average, 11 CD8+ LGL leukemia cell-specific high-confidence nonsynonymous somatic mutations were discovered in each patient. Interestingly, all patients had at least one mutation that affects either directly the STAT3-pathway (such as PTPRT) or T-cell activation (BCL11B, SLIT2 and NRP1). In all three patients, the STAT3 pathway was activated when studied by RNA expression or pSTAT3 analysis. Screening of the remaining 113 LGL leukemia patients did not reveal additional patients with same mutations. These novel mutations are potentially biologically relevant and represent rare genetic triggers for T-LGL leukemia, and are associated with similar disease phenotype as observed in patients with mutations in the STAT3 gene.

12.
Oncogene ; 32(41): 4892-902, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-23178494

ABSTRACT

Notch signaling is frequently hyperactivated in breast cancer, but how the enhanced signaling contributes to the tumor process is less well understood. In this report, we identify the proinflammatory cytokine interleukin-6 (IL-6) as a novel Notch target in breast tumor cells. Enhanced Notch signaling upregulated IL-6 expression, leading to activation of autocrine and paracrine Janus kinase/signal transducers and activators of transcription signaling. IL-6 upregulation was mediated by non-canonical Notch signaling, as it could be effectuated by a cytoplasmically localized Notch intracellular domain and was independent of the DNA-binding protein CSL. Instead, Notch-mediated IL-6 upregulation was controlled by two proteins in the nuclear factor (NF)-κB signaling cascade, IKKα and IKKß (inhibitor of nuclear factor kappa-B kinase subunit alpha and beta, respectively), as well as by p53. Activation of IL-6 by Notch required IKKα/IKKß function, but interestingly, did not engage canonical NF-κB signaling, in contrast to IL-6 activation by inflammatory agents such as lipopolysaccharide. With regard to p53 status, IL-6 expression was upregulated by Notch when p53 was mutated or lost, and restoring wild-type p53 into p53-mutated or -deficient cells abrogated the IL-6 upregulation. Furthermore, Notch-induced transcriptomes from p53 wild-type and -mutated breast tumor cell lines differed extensively, and for a subset of genes upregulated by Notch in a p53-mutant cell line, this upregulation was reduced by wild-type p53. In conclusion, we identify IL-6 as a novel non-canonical Notch target gene, and reveal roles for p53 and IKKα/IKKß in non-canonical Notch signaling in breast cancer and in the generation of cell context-dependent diversity in the Notch signaling output.


Subject(s)
Breast Neoplasms/pathology , I-kappa B Kinase/metabolism , Interleukin-6/metabolism , Janus Kinases/metabolism , Receptors, Notch/metabolism , STAT Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Autocrine Communication , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Interleukin-6/genetics , Macrophages/pathology , Paracrine Communication , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transcriptome , Up-Regulation
13.
Oncogene ; 31(39): 4266-78, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22249265

ABSTRACT

Protein phosphatase 2A (PP2A) is a critical human tumor-suppressor complex. A recently characterized PP2A inhibitor protein, namely cancerous inhibitor of PP2A (CIP2A), has been found to be overexpressed at a high frequency in most of the human cancer types. However, our understanding of gene expression programs regulated by CIP2A is almost absent. Moreover, clinical relevance of the CIP2A-regulated transcriptome has not been addressed thus far. Here, we report a high-confidence transcriptional signature regulated by CIP2A. Bioinformatic pathway analysis of the CIP2A signature revealed that CIP2A regulates several MYC-dependent and MYC-independent gene programs. With regard to MYC-independent signaling, JNK2 expression and transwell migration were inhibited by CIP2A depletion, whereas MYC depletion did not affect either of these phenotypes. Instead, depletion of either CIP2A or MYC inhibited cancer cell colony growth with statistically indistinguishable efficiency. Moreover, CIP2A depletion was shown to regulate the expression of several established MYC target genes, out of which most were MYC-repressed genes. CIP2A small-interfering RNA-elicited inhibition of colony growth or activation of MYC-repressed genes was reversed at large by concomitant PP2A inhibition. Finally, the CIP2A signature was shown to cluster with basal-type and human epidermal growth factor receptor (HER)2-positive (HER2+) breast cancer signatures. Accordingly, CIP2A protein expression was significantly associated with basal-like (P=0.0014) and HER2+ (P<0.0001) breast cancers. CIP2A expression also associated with MYC gene amplification (P<0.001). Taken together, identification of CIP2A-driven transcriptional signature, and especially novel MYC-independent signaling programs regulated by CIP2A, provides important resource for understanding CIP2A's role as a clinically relevant human oncoprotein. With regard to MYC, these results both validate CIP2A's role in regulating MYC-mediated gene expression and provide a plausible novel explanation for the high MYC activity in basal-like and HER2+ breast cancers.


Subject(s)
Autoantigens/metabolism , Breast Neoplasms/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Breast Neoplasms/drug therapy , Cell Proliferation/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Signaling Peptides and Proteins , Mitogen-Activated Protein Kinase 9/biosynthesis , Protein Phosphatase 2/metabolism , RNA, Small Interfering/pharmacology , Receptor, ErbB-2/analysis , Signal Transduction/drug effects , Tumor Cells, Cultured
14.
Br J Cancer ; 106(1): 99-106, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22215106

ABSTRACT

BACKGROUND: We have shown that a sodium ionophore monensin inhibits prostate cancer cell growth. A structurally related compound to monensin, salinomycin, was recently identified as a putative cancer stem cell inhibitor. METHODS: The growth inhibitory potential of salinomycin was studied in a panel of prostate cells. To get insights into the mechanism of action, a variety of assays such as gene expression and steroid profiling were performed in salinomycin-exposed prostate cancer cells. RESULTS: Salinomycin inhibited the growth of prostate cancer cells, but did not affect non-malignant prostate epithelial cells. Salinomycin impacted on prostate cancer stem cell functions as evidenced by reduced aldehyde dehydrogenase activity and the fraction of CD44(+) cells. Moreover, salinomycin reduced the expression of MYC, AR and ERG, induced oxidative stress as well as inhibited nuclear factor-κB activity and cell migration. Furthermore, profiling steroid metabolites revealed increased levels of oxidative stress-inducing steroids 7-ketocholesterol and aldosterone and decreased levels of antioxidative steroids progesterone and pregnenolone in salinomycin-exposed prostate cancer cells. CONCLUSION: Our results indicate that salinomycin inhibits prostate cancer cell growth and migration by reducing the expression of key prostate cancer oncogenes, inducing oxidative stress, decreasing the antioxidative capacity and cancer stem cell fraction.


Subject(s)
Cell Division/drug effects , Cell Movement/drug effects , Oxidative Stress/drug effects , Prostatic Neoplasms/pathology , Pyrans/pharmacology , Aldehyde Dehydrogenase/antagonists & inhibitors , Blotting, Western , Cell Line, Tumor , Humans , Male , NF-kappa B/metabolism , Niclosamide/pharmacology , Prostatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Receptors, Androgen/drug effects , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Terfenadine/pharmacology
15.
Oncogene ; 31(10): 1299-310, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-21785461

ABSTRACT

Malignant glioma is the most common brain tumor with 16,000 new cases diagnosed annually in the United States. We performed a systematic large-scale transcriptomics data mining study of 9783 tissue samples from the GeneSapiens database to systematically identify genes that are most glioma-specific. We searched for genes that were highly expressed in 322 glioblastoma multiforme tissue samples and 66 anaplastic astrocytomas as compared with 425 samples from histologically normal central nervous system. Transcription cofactor HES6 (hairy and enhancer of split 6) emerged as the most glioma-specific gene. Immunostaining of a tissue microarray showed HES6 expression in 335 (98.8%) out of the 339 glioma samples. HES6 was expressed in endothelial cells of the normal brain and glioma tissue. Recurrent grade 2 astrocytomas and grade 2 or 3 oligodendrogliomas showed higher levels of HES6 immunoreactivity than the corresponding primary tumors. High HES6 mRNA expression correlated with the proneural subtype that generally has a favorable outcome but is prone to recur. Functional studies suggested an important role for HES6 in supporting survival of glioma cells, as evidenced by reduction of cancer cell proliferation and migration after HES6 silencing. The biological role and consequences of HES6 silencing and overexpression was explored with genome-wide analyses, which implicated a role for HES6 in p53, c-myc and nuclear factor-κB transcriptional networks. We conclude that HES6 is important for glioma cell proliferation and migration, and may have a role in angiogenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Neoplasms/pathology , Cell Proliferation , Glioma/pathology , Repressor Proteins/genetics , Transcription, Genetic , Basic Helix-Loop-Helix Transcription Factors/analysis , Basic Helix-Loop-Helix Transcription Factors/physiology , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Movement , ErbB Receptors/genetics , Gene Dosage , Genes, myc , Glioma/genetics , Glioma/mortality , Humans , Receptor, Platelet-Derived Growth Factor alpha/genetics , Repressor Proteins/analysis , Repressor Proteins/physiology , Vascular Endothelial Growth Factor Receptor-2/genetics
16.
Oncogene ; 31(16): 2075-89, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-21996742

ABSTRACT

Normal prostate and some malignant prostate cancer (PrCa) cell lines undergo acinar differentiation and form spheroids in three-dimensional (3-D) organotypic culture. Acini formed by PC-3 and PC-3M, less pronounced also in other PrCa cell lines, spontaneously undergo an invasive switch, leading to the disintegration of epithelial structures and the basal lamina, and formation of invadopodia. This demonstrates the highly dynamic nature of epithelial plasticity, balancing epithelial-to-mesenchymal transition against metastable acinar differentiation. This study assessed the role of lipid metabolites on epithelial maturation. PC-3 cells completely failed to form acinar structures in delipidated serum. Adding back lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) rescued acinar morphogenesis and repressed invasion effectively. Blocking LPA receptor 1 (LPAR1) functions by siRNA (small interference RNA) or the specific LPAR1 inhibitor Ki16425 promoted invasion, while silencing of other G-protein-coupled receptors responsive to LPA or S1P mainly caused growth arrest or had no effects. The G-proteins Gα(12/13) and Gα(i) were identified as key mediators of LPA signalling via stimulation of RhoA and Rho kinases ROCK1 and 2, activating Rac1, while inhibition of adenylate cyclase and accumulation of cAMP may be secondary. Interfering with these pathways specifically impeded epithelial polarization in transformed cells. In contrast, blocking the same pathways in non-transformed, normal cells promoted differentiation. We conclude that LPA and LPAR1 effectively promote epithelial maturation and block invasion of PrCa cells in 3-D culture. The analysis of clinical transcriptome data confirmed reduced expression of LPAR1 in a subset of PrCa's. Our study demonstrates a metastasis-suppressor function for LPAR1 and Gα(12/13) signalling, regulating cell motility and invasion versus epithelial maturation.


Subject(s)
Cell Differentiation , Cell Movement , Lysophospholipids/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Sphingosine/analogs & derivatives , Cell Culture Techniques , Cell Line, Tumor , Epithelium/pathology , Epithelium/physiology , GTP-Binding Proteins/metabolism , Humans , Male , RNA Interference , Signal Transduction , Sphingosine/metabolism , Tumor Cells, Cultured
17.
Oncogene ; 31(16): 2090-100, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-21892209

ABSTRACT

Tumor-suppressor genes (TSGs) have been classically defined as genes whose loss of function in tumor cells contributes to the formation and/or maintenance of the tumor phenotype. TSGs containing nonsense mutations may not be expressed because of nonsense-mediated RNA decay (NMD). We combined inhibition of the NMD process, which clears transcripts that contain nonsense mutations, with the application of high-density single-nucleotide polymorphism arrays analysis to discriminate allelic content in order to identify candidate TSGs in five breast cancer cell lines. We identified ARID1A as a target of NMD in the T47D breast cancer cell line, likely as a consequence of a mutation in exon-9, which introduces a premature stop codon at position Q944. ARID1A encodes a human homolog of yeast SWI1, which is an integral member of the hSWI/SNF complex, an ATP-dependent, chromatin-remodeling, multiple-subunit enzyme. Although we did not find any somatic mutations in 11 breast tumors, which show DNA copy-number loss at the 1p36 locus adjacent to ARID1A, we show that low ARID1A RNA or nuclear protein expression is associated with more aggressive breast cancer phenotypes, such as high tumor grade, in two independent cohorts of over 200 human breast cancer cases each. We also found that low ARID1A nuclear expression becomes more prevalent during the later stages of breast tumor progression. Finally, we found that ARID1A re-expression in the T47D cell line results in significant inhibition of colony formation in soft agar. These results suggest that ARID1A may be a candidate TSG in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Genes, Tumor Suppressor , Nuclear Proteins/genetics , Transcription Factors/genetics , Cell Line, Tumor , Chromosomes, Human, Pair 1 , Codon, Nonsense , DNA Copy Number Variations , DNA-Binding Proteins , Female , Humans , RNA/metabolism , Transfection
18.
Oncogene ; 31(31): 3597-606, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22120710

ABSTRACT

Aneuploidy is frequently detected in solid tumors but the mechanisms regulating the generation of aneuploidy and their relevance in cancer initiation remain under debate and are incompletely characterized. Spatial and temporal regulation of integrin traffic is critical for cell migration and cytokinesis. Impaired integrin endocytosis, because of the loss of Rab21 small GTPase or mutations in the integrin ß-subunit cytoplasmic tail, induces failure of cytokinesis in vitro. Here, we describe that repeatedly failed cytokinesis, because of impaired traffic, is sufficient to trigger the generation of aneuploid cells, which display characteristics of oncogenic transformation in vitro and are tumorigenic in vivo. Furthermore, in an in vivo mouse xenograft model, non-transformed cells with impaired integrin traffic formed tumors with a long latency. More detailed investigation of these tumors revealed that the tumor cells were aneuploid. Therefore, abnormal integrin traffic was linked with generation of aneuploidy and cell transformation also in vivo. In human prostate and ovarian cancer samples, downregulation of Rab21 correlates with increased malignancy. Loss-of-function experiments demonstrate that long-term depletion of Rab21 is sufficient to induce chromosome number aberrations in normal human epithelial cells. These data are the first to demonstrate that impaired integrin traffic is sufficient to induce conversion of non-transformed cells to tumorigenic cells in vitro and in vivo.


Subject(s)
Aneuploidy , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cytokinesis , Integrins/metabolism , Animals , Breast/metabolism , Down-Regulation , Epithelial Cells/metabolism , Female , Humans , Male , Mice , Mice, Nude , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Transport/physiology , rab GTP-Binding Proteins/biosynthesis , rab GTP-Binding Proteins/genetics
19.
Oncogene ; 31(29): 3444-56, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22120715

ABSTRACT

Our understanding of key epigenetic regulators involved in specific biological processes and cancers is still incomplete, despite great progress in genome-wide studies of the epigenome. Here, we carried out a systematic, genome-wide analysis of the functional significance of 615 epigenetic proteins in prostate cancer (PrCa) cells. We used the high-content cell-spot microarray technology and siRNA silencing of PrCa cell lines for functional screening of cell proliferation, survival, androgen receptor (AR) expression, histone methylation and acetylation. Our study highlights subsets of epigenetic enzymes influencing different cancer cell phenotypes. Plant homeo domain (PHD) finger proteins have a key role in cell survival and histone methylation, whereas histone deacetylases were primarily involved in regulating AR expression. In contrast, JumonjiC-domain (JmjC) containing histone lysine demethylases (KDMs) mainly had an impact on cell proliferation. Our results show that the KDMs JARID1B, PHF8, KDM3A, KDM3B and KDM4A were highly expressed in clinical PrCa samples. The PHD-finger protein 8 (PHF8), a transcriptional coactivator with both PHD- and JmjC-domains, was moderately to strongly expressed in 80% of clinical PrCa samples, whereas 76% of normal and benign samples were negative or only showed weak PHF8 expression. Strong PHF8 expression correlated significantly with high Gleason grade and was borderline significant for poor prognosis. The results of functional PHF8 knockdown implicate a role in cell migration and invasion, as shown by cell motility and 3-D invasion assays. Our study suggests that various cellular phenotypes are regulated by distinct subsets of epigenetic enzymes. Proteins interpreting and modifying histone methylation, such as JmjC-domain and particularly PHD-finger proteins like PHF8, are activated in subsets of PrCa's and promote cancer relevant phenotypes.


Subject(s)
Cell Movement/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Histone Demethylases/deficiency , Histone Demethylases/genetics , Prostatic Neoplasms/pathology , Transcription Factors/deficiency , Transcription Factors/genetics , Adult , Aged , Aged, 80 and over , Cell Proliferation , Histone Deacetylases/deficiency , Histone Deacetylases/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/deficiency , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Middle Aged , Neoplasm Invasiveness/genetics , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism
20.
Oncogene ; 28(44): 3926-36, 2009 Nov 05.
Article in English | MEDLINE | ID: mdl-19684618

ABSTRACT

Predicting the impact of microRNAs (miRNAs) on target proteins is challenging because of their different regulatory effects at the transcriptional and translational levels. In this study, we applied a novel protein lysate microarray (LMA) technology to systematically monitor for target protein levels after high-throughput transfections of 319 pre-miRs into breast cancer cells. We identified 21 miRNAs that downregulated the estrogen receptor-alpha (ERalpha), as validated by western blotting and quantitative real time-PCR, and by demonstrating the inhibition of estrogen-stimulated cell growth. Five potent ERalpha-regulating miRNAs, miR-18a, miR-18b, miR-193b, miR-206 and miR-302c, were confirmed to directly target ERalpha in 3'-untranslated region reporter assays. The gene expression signature that they repressed highly overlapped with that of a small interfering RNA against ERalpha, and across all the signatures tested, was most closely associated with the repression of known estrogen-induced genes. Furthermore, miR-18a and miR-18b showed higher levels of expression in ERalpha-negative as compared with ERalpha-positive clinical tumors. In summary, we present systematic and direct functional evidence of miRNAs inhibiting ERalpha signaling in breast cancer, and demonstrate the high-throughput LMA technology as a novel, powerful technique in determining the relative impact of various miRNAs on key target proteins and associated cellular processes and pathways.


Subject(s)
Down-Regulation , Estrogen Receptor alpha/biosynthesis , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , RNA, Neoplasm/metabolism , Signal Transduction , 3' Untranslated Regions , Breast Neoplasms , Cell Line, Tumor , Cell-Free System/metabolism , Female , Humans , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL