Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Sci Rep ; 11(1): 11023, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34040017

ABSTRACT

BRAF inhibitors (BRAFi) selectively target oncogenic BRAFV600E/K and are effective in 80% of advanced cutaneous malignant melanoma cases carrying the V600 mutation. However, the development of drug resistance limits their clinical efficacy. Better characterization of the underlying molecular processes is needed to further improve treatments. We previously demonstrated that transcription of PTEN is negatively regulated by the PTEN pseudogene antisense RNA, PTENP1-AS, and here we investigated the impact of this transcript on clinical outcome and BRAFi resistance in melanoma. We observed that increased expression levels of PTENP1-AS in BRAFi resistant cells associated with enrichment of EZH2 and H3K27me3 at the PTEN promoter, consequently reducing the expression levels of PTEN. Further, we showed that targeting of the PTENP1-AS transcript sensitized resistant cells to BRAFi treatment and that high expression of PTENP1-AS in stage III melanoma correlated with poor survival. Collectively, the data presented here show that PTENP1-AS is a promising target for re-sensitizing cells to BRAFi and also a possible prognostic marker for clinical outcome in stage III melanoma.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Skin Neoplasms , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Vemurafenib/pharmacology , Melanoma, Cutaneous Malignant
2.
Melanoma Res ; 30(5): 443-454, 2020 10.
Article in English | MEDLINE | ID: mdl-32467529

ABSTRACT

Introduction of targeted therapy in the treatment of metastatic cutaneous malignant melanoma (CMM) has improved clinical outcome during the last years. However, only in a subset of the CMM patients, this will lead to long-term effects. CEBPB is a transcription factor that has been implicated in various physiological and pathological processes, including cancer development. We have investigated its prognostic impact on CMM and unexpectedly found that higher CEBPB mRNA levels correlated with a longer overall survival. Furthermore, in a small cohort of patients with metastatic CMM treated with BRAF-inhibitors, higher levels of CEBPB mRNA expression in the tumor cells prior treatment correlated to a longer progression-free survival. We have characterized an overlapping antisense transcript, CEBPB-AS1, with the aim to investigate the regulation of CEBPB expression in CMM and its impact on BRAF-inhibitor sensitivity. We demonstrated that silencing of CEBPB-AS1 resulted in epigenetic modifications in the CEBPB promoter and in increased CEBPB mRNA and protein levels, inhibited proliferation and partially resensitized BRAF-inhibitor resistant CMM cells to this drug-induced apoptosis. Our data suggest that targeting CEBPB-AS1 may represent a valuable tool to sensitize CMM cells to the BRAF-inhibitor-based therapies.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Melanoma/drug therapy , RNA, Antisense/genetics , Vemurafenib/pharmacology , Adult , Aged , Aged, 80 and over , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Male , Melanoma/genetics , Melanoma/metabolism , Middle Aged , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , RNA, Antisense/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism
3.
EMBO Mol Med ; 12(3): e10419, 2020 03 06.
Article in English | MEDLINE | ID: mdl-31950591

ABSTRACT

The deoxycytidine analogue cytarabine (ara-C) remains the backbone treatment of acute myeloid leukaemia (AML) as well as other haematological and lymphoid malignancies, but must be combined with other chemotherapeutics to achieve cure. Yet, the underlying mechanism dictating synergistic efficacy of combination chemotherapy remains largely unknown. The dNTPase SAMHD1, which regulates dNTP homoeostasis antagonistically to ribonucleotide reductase (RNR), limits ara-C efficacy by hydrolysing the active triphosphate metabolite ara-CTP. Here, we report that clinically used inhibitors of RNR, such as gemcitabine and hydroxyurea, overcome the SAMHD1-mediated barrier to ara-C efficacy in primary blasts and mouse models of AML, displaying SAMHD1-dependent synergy with ara-C. We present evidence that this is mediated by dNTP pool imbalances leading to allosteric reduction of SAMHD1 ara-CTPase activity. Thus, SAMHD1 constitutes a novel biomarker for combination therapies of ara-C and RNR inhibitors with immediate consequences for clinical practice to improve treatment of AML.


Subject(s)
Cytarabine/pharmacology , Leukemia, Myeloid, Acute , Pyrophosphatases/metabolism , Ribonucleotide Reductases/antagonists & inhibitors , SAM Domain and HD Domain-Containing Protein 1/metabolism , Animals , Arabinofuranosylcytosine Triphosphate/metabolism , Mice
4.
J Pharm Biomed Anal ; 160: 80-88, 2018 Oct 25.
Article in English | MEDLINE | ID: mdl-30086509

ABSTRACT

STAT3 protein is an established target for the development of new cancer therapeutic agents. Despite lacking a traditional binding site for small molecule inhibitors, many STAT3 inhibitors have been identified and explored for their anti-cancer activity. Because STAT3 signaling is mediated by protein-protein interactions, indirect methods are often employed to determine if proposed STAT3 inhibitors bind to STAT3 protein. While established STAT3 inhibition assays (such as the fluorescence polarization assay, electrophoretic mobility shift assay and ELISAs) have been used to identify novel inhibitors of STAT3 signaling, methods that directly assess STAT3 protein-inhibitor interactions could facilitate the development of novel inhibitors. In this context, we herein report new STAT3 binding assays based on differential scanning fluorimetry (DSF) and differential scanning light scattering (DSLS) to characterize interactions between STAT3 protein and inhibitors. Several peptide and small molecule STAT3 inhibitors have been evaluated, and new insight into how these compounds may interact with STAT3 is provided.


Subject(s)
Drug Development/methods , Fluorometry/methods , Peptides/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Binding Sites , Cyclic S-Oxides/chemistry , Cyclic S-Oxides/pharmacology , High-Throughput Screening Assays/methods , Light , Peptides/chemistry , Protein Binding , Protein Domains , Protein Stability , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/isolation & purification , Scattering, Radiation , Temperature
5.
Cell Death Dis ; 9(9): 846, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30154400

ABSTRACT

Glucocorticoids (GCs) are metabolic hormones with immunosuppressive effects that have proven effective drugs against childhood acute lymphoblastic leukemia (ALL). Yet, the role of metabolic reprogramming in GC-induced ALL cell death is poorly understood. GCs efficiently block glucose uptake and metabolism in ALL cells, but this does not fully explain the observed induction of autophagy and cell death. Here, we have performed parallel time-course proteomics, metabolomics, and isotope-tracing studies to examine in detail the metabolic effects of GCs on ALL cells. We observed metabolic events associated with growth arrest, autophagy, and catabolism prior to onset of apoptosis: nucleotide de novo synthesis was reduced, while certain nucleobases accumulated; polyamine synthesis was inhibited; and phosphatidylcholine synthesis was induced. GCs suppressed not only glycolysis but also entry of both glucose and glutamine into the TCA cycle. In contrast, expression of glutamine-ammonia ligase (GLUL) and cellular glutamine content was robustly increased by GC treatment, suggesting induction of glutamine synthesis, similar to nutrient-starved muscle. Modulating medium glutamine and dimethyl-α-ketoglutarate (dm-αkg) to favor glutamine synthesis reduced autophagosome content of ALL cells, and dm-αkg also rescued cell viability. These data suggest that glutamine synthesis affects autophagy and possibly onset of cell death in response to GCs, which should be further explored to understand mechanism of action and possible sources of resistance.


Subject(s)
Glucocorticoids/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Citric Acid Cycle/drug effects , Glutamine/metabolism , Glycolysis/drug effects , Humans
6.
Cancer Lett ; 435: 32-43, 2018 10 28.
Article in English | MEDLINE | ID: mdl-30055290

ABSTRACT

Resistance to chemotherapy is a challenging problem for treatment of cancer patients and autophagy has been shown to mediate development of resistance. In this study we systematically screened a library of 306 known anti-cancer drugs for their ability to induce autophagy using a cell-based assay. 114 of the drugs were classified as autophagy inducers; for 16 drugs, the cytotoxicity was potentiated by siRNA-mediated knock-down of Atg7 and Vps34. These drugs were further evaluated in breast cancer cell lines for autophagy induction, and two tyrosine kinase inhibitors, Sunitinib and Erlotinib, were selected for further studies. For the pharmacological inhibition of autophagy, we have characterized here a novel highly potent selective inhibitor of Vps34, SB02024. SB02024 blocked autophagy in vitro and reduced xenograft growth of two breast cancer cell lines, MDA-MB-231 and MCF-7, in vivo. Vps34 inhibitor significantly potentiated cytotoxicity of Sunitinib and Erlotinib in MCF-7 and MDA-MB-231 in vitro in monolayer cultures and when grown as multicellular spheroids. Our data suggests that inhibition of autophagy significantly improves sensitivity to Sunitinib and Erlotinib and that Vps34 is a promising therapeutic target for combination strategies in breast cancer.


Subject(s)
Autophagy/drug effects , Breast Neoplasms/drug therapy , Class III Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Xenograft Model Antitumor Assays/methods , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Class III Phosphatidylinositol 3-Kinases/metabolism , Drug Screening Assays, Antitumor/methods , Humans , MCF-7 Cells , Mice, Inbred NOD , Mice, SCID , Protein Kinase Inhibitors/pharmacology , Sunitinib/pharmacology
7.
Cell Death Dis ; 9(7): 736, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29970884

ABSTRACT

The microRNA-34a is a well-studied tumor suppressor microRNA (miRNA) and a direct downstream target of TP53 with roles in several pathways associated with oncogenesis, such as proliferation, cellular growth, and differentiation. Due to its broad tumor suppressive activity, it is not surprising that miR34a expression is altered in a wide variety of solid tumors and hematological malignancies. However, the mechanisms by which miR34a is regulated in these cancers is largely unknown. In this study, we find that a long noncoding RNA transcribed antisense to the miR34a host gene, is critical for miR34a expression and mediation of its cellular functions in multiple types of human cancer. We name this long noncoding RNA lncTAM34a, and characterize its ability to facilitate miR34a expression under different types of cellular stress in both TP53-deficient and wild-type settings.


Subject(s)
MicroRNAs/metabolism , RNA, Antisense/physiology , Blotting, Western , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Chromatin Immunoprecipitation , Computational Biology , DNA Damage/genetics , DNA Damage/physiology , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Genes, Tumor Suppressor/physiology , Humans , MicroRNAs/genetics , Promoter Regions, Genetic/genetics , RNA, Antisense/genetics , Tandem Mass Spectrometry
8.
Bioinformatics ; 33(19): 3126-3128, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28957498

ABSTRACT

SUMMARY: Multi-dimensional data generated via high-throughput experiments is increasingly used in conjunction with dimensionality reduction methods to ascertain if resulting separations of the data correspond with known classes. This is particularly useful to determine if a subset of the variables, e.g. genes in a specific pathway, alone can separate samples into these established classes. Despite this, the evaluation of class separations is often subjective and performed via visualization. Here we present the ClusterSignificance package; a set of tools designed to assess the statistical significance of class separations downstream of dimensionality reduction algorithms. In addition, we demonstrate the design and utility of the ClusterSignificance package and utilize it to determine the importance of long non-coding RNA expression in the identity of multiple hematological malignancies. AVAILABILITY AND IMPLEMENTATION: ClusterSignificance is an R package available via Bioconductor (https://bioconductor.org/packages/ClusterSignificance) under GPL-3. CONTACT: dan.grander@ki.se. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Gene Expression Profiling/methods , Software , Algorithms , Cluster Analysis , Data Interpretation, Statistical , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Humans , RNA, Long Noncoding/metabolism
9.
Proc Natl Acad Sci U S A ; 114(37): 9942-9947, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28847966

ABSTRACT

RNA has been found to interact with chromatin and modulate gene transcription. In human cells, little is known about how long noncoding RNAs (lncRNAs) interact with target loci in the context of chromatin. We find here, using the phosphatase and tensin homolog (PTEN) pseudogene as a model system, that antisense lncRNAs interact first with a 5' UTR-containing promoter-spanning transcript, which is then followed by the recruitment of DNA methyltransferase 3a (DNMT3a), ultimately resulting in the transcriptional and epigenetic control of gene expression. Moreover, we find that the lncRNA and promoter-spanning transcript interaction are based on a combination of structural and sequence components of the antisense lncRNA. These observations suggest, on the basis of this one example, that evolutionary pressures may be placed on RNA structure more so than sequence conservation. Collectively, the observations presented here suggest a much more complex and vibrant RNA regulatory world may be operative in the regulation of gene expression.


Subject(s)
PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/physiology , Chromatin/genetics , Chromatin Assembly and Disassembly , DNA Methyltransferase 3A , Exons , HEK293 Cells , Humans , Molecular Dynamics Simulation , Nucleic Acid Conformation , Promoter Regions, Genetic/genetics , Pseudogenes , Regulatory Elements, Transcriptional/genetics , Regulatory Elements, Transcriptional/physiology , Sequence Analysis, RNA/methods , Sequence Homology
10.
J Control Release ; 261: 199-206, 2017 09 10.
Article in English | MEDLINE | ID: mdl-28684168

ABSTRACT

Epidemiological studies of childhood leukemia survivors reveal an alarmingly high incidence of chronic health disabilities after treatment, therefore, more specific therapies need to be developed. Polo-like kinase 1 (Plk1) is a key player in mitosis and a target for drug development as it is upregulated in multiple cancer types. Small molecules targeting Plk1 are mainly ATP-competitors and, therefore, are known to elicit side effects due to lack of specificity. RNA interference (RNAi) is known for its high catalytic activity and target selectivity; however, the biggest barrier for its introduction into clinical use is its delivery. RNAi prodrugs are modified, self-delivering short interfering Ribonucleic Neutrals (siRNNs), cleaved by cytoplasmic enzymes into short interfering Ribonucleic Acids (siRNAs) once inside cells. In this study we aimed to investigate the potential of siRNNs as therapeutic tools in T-acute lymphoblastic leukemia (T-ALL) using T-ALL cell lines and patient-derived samples. We demonstrate for the first time that RNAi prodrugs (siRNNs) targeting Plk1, can enter pediatric T-ALL patient cells without a transfection reagent and induce Plk1 knockdown on both protein and mRNA levels resulting in G2/M-arrest and apoptosis. We also show that siRNNs targeting Plk1 generate less toxicity in normal cells compared to the small molecule Plk1 inhibitor, BI6727, suggesting a potentially good therapeutic index.


Subject(s)
Cell Cycle Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Small Interfering/administration & dosage , Apoptosis/genetics , Cell Line, Tumor , Child , Drug Delivery Systems , G2 Phase Cell Cycle Checkpoints/genetics , Gene Knockdown Techniques , Gene Silencing , Humans , M Phase Cell Cycle Checkpoints/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Prodrugs , Pteridines/pharmacology , Pteridines/toxicity , RNA, Messenger/genetics , RNA, Small Interfering/toxicity , Polo-Like Kinase 1
11.
PLoS One ; 12(6): e0178844, 2017.
Article in English | MEDLINE | ID: mdl-28636670

ABSTRACT

Activation of Signal Transducer and Activator of Transcription 3 (STAT3) has been linked to several processes that are critical for oncogenic transformation, cancer progression, cancer cell proliferation, survival, drug resistance and metastasis. Inhibition of STAT3 signaling has shown a striking ability to inhibit cancer cell growth and therefore, STAT3 has become a promising target for anti-cancer drug development. The aim of this study was to identify novel inhibitors of STAT-dependent gene transcription. A cellular reporter-based system for monitoring STAT3 transcriptional activity was developed which was suitable for high-throughput screening (Z' = 0,8). This system was used to screen a library of 28,000 compounds (the ENAMINE Drug-Like Diversity Set). Following counter-screenings and toxicity studies, we identified four hit compounds that were subjected to detailed biological characterization. Of the four hits, KI16 stood out as the most promising compound, inhibiting STAT3 phosphorylation and transcriptional activity in response to IL6 stimulation. In silico docking studies showed that KI16 had favorable interactions with the STAT3 SH2 domain, however, no inhibitory activity could be observed in the STAT3 fluorescence polarization assay. KI16 inhibited cell viability preferentially in STAT3-dependent cell lines. Taken together, using a targeted, cell-based approach, novel inhibitors of STAT-driven transcriptional activity were discovered which are interesting leads to pursue further for the development of anti-cancer therapeutic agents.


Subject(s)
Antineoplastic Agents/pharmacology , High-Throughput Screening Assays/methods , STAT3 Transcription Factor/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Signal Transduction , Small Molecule Libraries/chemistry , Tumor Cells, Cultured
12.
Cancer Immunol Res ; 5(8): 654-665, 2017 08.
Article in English | MEDLINE | ID: mdl-28637877

ABSTRACT

Manipulation of human natural killer (NK) cell repertoires promises more effective strategies for NK cell-based cancer immunotherapy. A subset of highly differentiated NK cells, termed adaptive NK cells, expands naturally in vivo in response to human cytomegalovirus (HCMV) infection, carries unique repertoires of inhibitory killer cell immunoglobulin-like receptors (KIR), and displays strong cytotoxicity against tumor cells. Here, we established a robust and scalable protocol for ex vivo generation and expansion of adaptive NK cells for cell therapy against pediatric acute lymphoblastic leukemia (ALL). Culture of polyclonal NK cells together with feeder cells expressing HLA-E, the ligand for the activating NKG2C receptor, led to selective expansion of adaptive NK cells with enhanced alloreactivity against HLA-mismatched targets. The ex vivo expanded adaptive NK cells gradually obtained a more differentiated phenotype and were specific and highly efficient killers of allogeneic pediatric T- and precursor B-cell acute lymphoblastic leukemia (ALL) blasts, previously shown to be refractory to killing by autologous NK cells and the NK-cell line NK92 currently in clinical testing. Selective expansion of NK cells that express one single inhibitory KIR for self-HLA class I would allow exploitation of the full potential of NK-cell alloreactivity in cancer immunotherapy. In summary, our data suggest that adaptive NK cells may hold utility for therapy of refractory ALL, either as a bridge to transplant or for patients that lack stem cell donors. Cancer Immunol Res; 5(8); 654-65. ©2017 AACR.


Subject(s)
Immunotherapy , Killer Cells, Natural/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, KIR/immunology , Adaptive Immunity , Cell Line, Tumor , Cell- and Tissue-Based Therapy , Child , Cytomegalovirus/immunology , Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/therapeutic use , Humans , NK Cell Lectin-Like Receptor Subfamily C/immunology , NK Cell Lectin-Like Receptor Subfamily C/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptors, KIR/therapeutic use , HLA-E Antigens
13.
Nat Med ; 23(2): 256-263, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28067901

ABSTRACT

The cytostatic deoxycytidine analog cytarabine (ara-C) is the most active agent available against acute myelogenous leukemia (AML). Together with anthracyclines, ara-C forms the backbone of AML treatment for children and adults. In AML, both the cytotoxicity of ara-C in vitro and the clinical response to ara-C therapy are correlated with the ability of AML blasts to accumulate the active metabolite ara-C triphosphate (ara-CTP), which causes DNA damage through perturbation of DNA synthesis. Differences in expression levels of known transporters or metabolic enzymes relevant to ara-C only partially account for patient-specific differential ara-CTP accumulation in AML blasts and response to ara-C treatment. Here we demonstrate that the deoxynucleoside triphosphate (dNTP) triphosphohydrolase SAM domain and HD domain 1 (SAMHD1) promotes the detoxification of intracellular ara-CTP pools. Recombinant SAMHD1 exhibited ara-CTPase activity in vitro, and cells in which SAMHD1 expression was transiently reduced by treatment with the simian immunodeficiency virus (SIV) protein Vpx were dramatically more sensitive to ara-C-induced cytotoxicity. CRISPR-Cas9-mediated disruption of the gene encoding SAMHD1 sensitized cells to ara-C, and this sensitivity could be abrogated by ectopic expression of wild-type (WT), but not dNTPase-deficient, SAMHD1. Mouse models of AML lacking SAMHD1 were hypersensitive to ara-C, and treatment ex vivo with Vpx sensitized primary patient-derived AML blasts to ara-C. Finally, we identified SAMHD1 as a risk factor in cohorts of both pediatric and adult patients with de novo AML who received ara-C treatment. Thus, SAMHD1 expression levels dictate patient sensitivity to ara-C, providing proof-of-concept that the targeting of SAMHD1 by Vpx could be an attractive therapeutic strategy for potentiating ara-C efficacy in hematological malignancies.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cytarabine/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Monomeric GTP-Binding Proteins/drug effects , Viral Regulatory and Accessory Proteins/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antimetabolites, Antineoplastic/therapeutic use , Arabinofuranosylcytosine Triphosphate/metabolism , Child , Child, Preschool , Cytarabine/therapeutic use , Disease Models, Animal , Female , Humans , In Vitro Techniques , Infant , Leukemia, Myeloid, Acute/metabolism , Male , Mice , Molecular Targeted Therapy , Monomeric GTP-Binding Proteins/metabolism , Prognosis , SAM Domain and HD Domain-Containing Protein 1
14.
Oncotarget ; 7(39): 64071-64088, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27590521

ABSTRACT

To characterize the mutational patterns of acute lymphoblastic leukemia (ALL) we performed deep next generation sequencing of 872 cancer genes in 172 diagnostic and 24 relapse samples from 172 pediatric ALL patients. We found an overall greater mutational burden and more driver mutations in T-cell ALL (T-ALL) patients compared to B-cell precursor ALL (BCP-ALL) patients. In addition, the majority of the mutations in T-ALL had occurred in the original leukemic clone, while most of the mutations in BCP-ALL were subclonal. BCP-ALL patients carrying any of the recurrent translocations ETV6-RUNX1, BCR-ABL or TCF3-PBX1 harbored few mutations in driver genes compared to other BCP-ALL patients. Specifically in BCP-ALL, we identified ATRX as a novel putative driver gene and uncovered an association between somatic mutations in the Notch signaling pathway at ALL diagnosis and increased risk of relapse. Furthermore, we identified EP300, ARID1A and SH2B3 as relapse-associated genes. The genes highlighted in our study were frequently involved in epigenetic regulation, associated with germline susceptibility to ALL, and present in minor subclones at diagnosis that became dominant at relapse. We observed a high degree of clonal heterogeneity and evolution between diagnosis and relapse in both BCP-ALL and T-ALL, which could have implications for the treatment efficiency.


Subject(s)
High-Throughput Nucleotide Sequencing , Mutation , Neoplasm Recurrence, Local/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adaptor Proteins, Signal Transducing , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , DNA-Binding Proteins , E1A-Associated p300 Protein/genetics , Epigenesis, Genetic , Humans , Immunophenotyping , Infant , Intracellular Signaling Peptides and Proteins , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proteins/genetics , Recurrence , Remission Induction , Sequence Analysis, DNA , Transcription Factors/genetics , Translocation, Genetic
15.
J Invest Dermatol ; 136(12): 2485-2494, 2016 12.
Article in English | MEDLINE | ID: mdl-27452220

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC) is the second most common cancer and a leading cause of cancer mortality among solid organ transplant recipients. MicroRNAs (miR) are short RNAs that regulate gene expression and cellular functions. Here, we show a negative correlation between miR-203 expression and the differentiation grade of cSCC. Functionally, miR-203 suppressed cell proliferation, cell motility, and the angiogenesis-inducing capacity of cSCC cells in vitro and reduced xenograft tumor volume and angiogenesis in vivo. Transcriptomic analysis of cSCC cells with ectopic overexpression of miR-203 showed dramatic changes in gene networks related to cell cycle and proliferation. Transcription factor enrichment analysis identified c-MYC as a hub of miR-203-induced transcriptomic changes in squamous cell carcinoma. We identified c-MYC as a direct target of miR-203. Overexpression of c-MYC in rescue experiments reversed miR-203-induced growth arrest in cSCC, which highlights the importance of c-MYC within the miR-203-regulated gene network. Together, miR-203 acts as a tumor suppressor in cSCC, and its low expression can be a marker for poorly differentiated tumors. Restoration of miR-203 expression may provide a therapeutic benefit, particularly in poorly differentiated cSCC.


Subject(s)
Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Neoplastic , Genes, myc , MicroRNAs/genetics , Skin Neoplasms/pathology , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/genetics , Female , Gene Expression Profiling , Genes, Tumor Suppressor , Humans , Male , Neoplasm Invasiveness/pathology , Neoplasm Staging , Neovascularization, Pathologic/genetics , Sampling Studies , Sensitivity and Specificity , Skin Neoplasms/genetics , Up-Regulation
16.
Oncotarget ; 7(24): 35703-35723, 2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27248168

ABSTRACT

Sustained autophagy contributes to the metabolic adaptation of cancer cells to hypoxic and acidic microenvironments. Since cells in such environments are resistant to conventional cytotoxic drugs, inhibition of autophagy represents a promising therapeutic strategy in clinical oncology. We previously reported that the efficacy of hydroxychloroquine (HCQ), an autophagy inhibitor under clinical investigation is strongly impaired in acidic tumor environments, due to poor uptake of the drug, a phenomenon widely associated with drug resistance towards many weak bases. In this study we identified salinomycin (SAL) as a potent inhibitor of autophagy and cytotoxic agent effective on several cancer cell lines under conditions of transient and chronic acidosis. Since SAL has been reported to specifically target cancer-stem cells (CSC), we used an established model of breast CSC and CSC derived from breast cancer patients to examine whether this specificity may be associated with autophagy inhibition. We indeed found that CSC-like cells are more sensitive to autophagy inhibition compared to cells not expressing CSC markers. We also report that the ability of SAL to inhibit mammosphere formation from CSC-like cells was dramatically enhanced in acidic conditions. We propose that the development and use of clinically suitable SAL derivatives may result in improved autophagy inhibition in cancer cells and CSC in the acidic tumor microenvironment and lead to clinical benefits.


Subject(s)
Acidosis/physiopathology , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Breast Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Pyrans/pharmacology , Antineoplastic Agents/therapeutic use , Biopsy , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cell Survival , Female , Humans , Pyrans/therapeutic use , Spheroids, Cellular/drug effects , Spheroids, Cellular/physiology , Tumor Microenvironment/physiology , Tumor Stem Cell Assay
17.
Curr Top Microbiol Immunol ; 394: 111-26, 2016.
Article in English | MEDLINE | ID: mdl-25982975

ABSTRACT

Pseudogenes have for long been considered as non-functional relics littering the human genome. Only now, it is becoming apparent that many pseudogenes are transcribed into long noncoding RNAs, some with proven biological functions. Here, we review the current knowledge of pseudogenes and their widespread functional properties with an emphasis on pseudogenes that have been functionally investigated in greater detail. Pseudogenes are emerging as a novel class of long noncoding RNAs functioning, for example, through microRNA sponging and chromatin remodeling. The examples discussed herein underline that pseudogene-encoded RNAs are important regulatory molecules involved in diseases such as cancer.


Subject(s)
Pseudogenes/physiology , HMGA1a Protein/genetics , Humans , Neoplasms/genetics , Octamer Transcription Factor-3/genetics , PTEN Phosphohydrolase/genetics , RNA, Long Noncoding/physiology
18.
Oncotarget ; 6(35): 37066-82, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26416459

ABSTRACT

Autophagy is one of the main cytoprotective mechanisms that cancer cells deploy to withstand the cytotoxic stress and survive the lethal damage induced by anti-cancer drugs. However, under specific conditions, autophagy may, directly or indirectly, induce cell death. In our study, treatment of the Atg5-deficient DU145 prostate cancer cells, with the multi-tyrosine kinase inhibitor, sorafenib, induces mitochondrial damage, autophagy and cell death. Molecular inhibition of autophagy by silencing ULK1 and Beclin1 rescues DU145 cells from cell death indicating that, in this setting, autophagy promotes cell death. Re-expression of Atg5 restores the lipidation of LC3 and rescues DU145 and MEF atg5-/- cells from sorafenib-induced cell death. Despite the lack of Atg5 expression and LC3 lipidation, DU145 cells form autophagosomes as demonstrated by transmission and immuno-electron microscopy, and the formation of LC3 positive foci. However, the lack of cellular content in the autophagosomes, the accumulation of long-lived proteins, the presence of GFP-RFP-LC3 positive foci and the accumulated p62 protein levels indicate that these autophagosomes may not be fully functional. DU145 cells treated with sorafenib undergo a caspase-independent cell death that is inhibited by the RIPK1 inhibitor, necrostatin-1. Furthermore, treatment with sorafenib induces the interaction of RIPK1 with p62, as demonstrated by immunoprecipitation and a proximity ligation assay. Silencing of p62 decreases the RIPK1 protein levels and renders necrostatin-1 ineffective in blocking sorafenib-induced cell death. In summary, the formation of Atg5-deficient autophagosomes in response to sorafenib promotes the interaction of p62 with RIPK leading to cell death by necroptosis.


Subject(s)
Apoptosis/drug effects , Autophagy , Embryo, Mammalian/pathology , Fibroblasts/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Prostatic Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Autophagy-Related Protein 5 , Blotting, Western , Cells, Cultured , Drug Resistance, Neoplasm , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoprecipitation , Male , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/physiology , Necrosis , Niacinamide/pharmacology , Phagosomes/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Sorafenib , Tissue Array Analysis
19.
Int J Cancer ; 136(4): E51-61, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25156627

ABSTRACT

The mechanism of multicellular drug resistance, defined as the reduced efficacy of chemotherapeutic drugs in solid tumors is incompletely understood. Here we report that colon carcinoma cells cultured as 3D microtissues (spheroids) display dramatic increases in the expression of a subset of type I interferon-(IFN)-stimulated genes (ISGs). A similar gene signature was associated previously with resistance to radiation and chemotherapy, prompting us to examine the underlying biological mechanisms. Analysis of spheroids formed by different tumor cell lines and studies using knock-down of gene expression showed that cell crowding leads to the induction of IFN regulatory factor-9 (IRF9) which together with STAT2 and independently of IFNs, is necessary for ISG upregulation. Increased expression of IRF9 alone was sufficient to induce the ISG subset in monolayer cells and to confer increased resistance to clinically used cytotoxic drugs. Our data reveal a novel mechanism of regulation of a subset of ISGs, leading to drug resistance in solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics , Apoptosis , Cell Communication , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferons/physiology , STAT2 Transcription Factor/metabolism , Transcriptional Activation
20.
Hum Mutat ; 36(1): 118-28, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25355294

ABSTRACT

Genomic characterization of pediatric acute lymphoblastic leukemia (ALL) has identified distinct patterns of genes and pathways altered in patients with well-defined genetic aberrations. To extend the spectrum of known somatic variants in ALL, we performed whole genome and transcriptome sequencing of three B-cell precursor patients, of which one carried the t(12;21)ETV6-RUNX1 translocation and two lacked a known primary genetic aberration, and one T-ALL patient. We found that each patient had a unique genome, with a combination of well-known and previously undetected genomic aberrations. By targeted sequencing in 168 patients, we identified KMT2D and KIF1B as novel putative driver genes. We also identified a putative regulatory non-coding variant that coincided with overexpression of the growth factor MDK. Our results contribute to an increased understanding of the biological mechanisms that lead to ALL and suggest that regulatory variants may be more important for cancer development than recognized to date. The heterogeneity of the genetic aberrations in ALL renders whole genome sequencing particularly well suited for analysis of somatic variants in both research and diagnostic applications.


Subject(s)
DNA-Binding Proteins/genetics , High-Throughput Nucleotide Sequencing/methods , Kinesins/genetics , Mutation , Neoplasm Proteins/genetics , Nerve Growth Factors/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Child , Child, Preschool , Female , Genome, Human , Humans , Infant , Male , Midkine , Sequence Analysis, DNA/methods , Sequence Analysis, RNA/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...