Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cancer Res ; 82(24): 4586-4603, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36219398

ABSTRACT

Relapse is the leading cause of death in patients with medulloblastoma, the most common malignant pediatric brain tumor. A better understanding of the mechanisms underlying recurrence could lead to more effective therapies for targeting tumor relapses. Here, we observed that SOX9, a transcription factor and stem cell/glial fate marker, is limited to rare, quiescent cells in high-risk medulloblastoma with MYC amplification. In paired primary-recurrent patient samples, SOX9-positive cells accumulated in medulloblastoma relapses. SOX9 expression anti-correlated with MYC expression in murine and human medulloblastoma cells. However, SOX9-positive cells were plastic and could give rise to a MYC high state. To follow relapse at the single-cell level, an inducible dual Tet model of medulloblastoma was developed, in which MYC expression was redirected in vivo from treatment-sensitive bulk cells to dormant SOX9-positive cells using doxycycline treatment. SOX9 was essential for relapse initiation and depended on suppression of MYC activity to promote therapy resistance, epithelial-mesenchymal transition, and immune escape. p53 and DNA repair pathways were downregulated in recurrent tumors, whereas MGMT was upregulated. Recurrent tumor cells were found to be sensitive to treatment with an MGMT inhibitor and doxorubicin. These findings suggest that recurrence-specific targeting coupled with DNA repair inhibition comprises a potential therapeutic strategy in patients affected by medulloblastoma relapse. SIGNIFICANCE: SOX9 facilitates therapy escape and recurrence in medulloblastoma via temporal inhibition of MYC/MYCN genes, revealing a strategy to specifically target SOX9-positive cells to prevent tumor relapse.


Subject(s)
Brain Neoplasms , Cerebellar Neoplasms , Medulloblastoma , Animals , Humans , Mice , Cerebellar Neoplasms/pathology , Medulloblastoma/pathology , Neoplasm Recurrence, Local/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcription Factors/metabolism
2.
Epigenomes ; 5(4)2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34968247

ABSTRACT

Aberrant DNA methylation, dysregulation of chromatin-modifying enzymes, and microRNAs (miRNAs) play a crucial role in haematological malignancies. These epimutations, with an impact on chromatin accessibility and transcriptional output, are often associated with genomic instability and the emergence of drug resistance, disease progression, and poor survival. In order to exert their functions, epigenetic enzymes utilize cellular metabolites as co-factors and are highly dependent on their availability. By affecting the expression of metabolic enzymes, epigenetic modifiers may aid the generation of metabolite signatures that could be utilized as targets and biomarkers in cancer. This interdependency remains often neglected and poorly represented in studies, despite well-established methods to study the cellular metabolome. This review critically summarizes the current knowledge in the field to provide an integral picture of the interplay between epigenomic alterations and the cellular metabolome in haematological malignancies. Our recent findings defining a distinct metabolic signature upon response to enhancer of zeste homolog 2 (EZH2) inhibition in multiple myeloma (MM) highlight how a shift of preferred metabolic pathways may potentiate novel treatments. The suggested link between the epigenome and the metabolome in haematopoietic tumours holds promise for the use of metabolic signatures as possible biomarkers of response to treatment.

3.
Cell Death Dis ; 12(2): 167, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33579905

ABSTRACT

Multiple myeloma (MM) is a heterogeneous haematological disease that remains clinically challenging. Increased activity of the epigenetic silencer EZH2 is a common feature in patients with poor prognosis. Previous findings have demonstrated that metabolic profiles can be sensitive markers for response to treatment in cancer. While EZH2 inhibition (EZH2i) has proven efficient in inducing cell death in a number of human MM cell lines, we hereby identified a subset of cell lines that despite a global loss of H3K27me3, remains viable after EZH2i. By coupling liquid chromatography-mass spectrometry with gene and miRNA expression profiling, we found that sensitivity to EZH2i correlated with distinct metabolic signatures resulting from a dysregulation of genes involved in methionine cycling. Specifically, EZH2i resulted in a miRNA-mediated downregulation of methionine cycling-associated genes in responsive cells. This induced metabolite accumulation and DNA damage, leading to G2 arrest and apoptosis. Altogether, we unveiled that sensitivity to EZH2i in human MM cell lines is associated with a specific metabolic and gene expression profile post-treatment.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Methylation/drug effects , Drug Resistance, Neoplasm , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Metabolome , Methionine/metabolism , Multiple Myeloma/drug therapy , Pyridones/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Transcriptome
4.
Semin Cancer Biol ; 51: 101-115, 2018 08.
Article in English | MEDLINE | ID: mdl-28962927

ABSTRACT

Multiple myeloma (MM) is a tumor of antibody producing plasmablasts/plasma cells that resides within the bone marrow (BM). In addition to the well-established role of genetic lesions and tumor-microenvironment interactions in the development of MM, deregulated epigenetic mechanisms are emerging as important in MM pathogenesis. Recently, MM sequencing and expression projects have revealed that mutations and copy number variations as well as deregulation in the expression of epigenetic modifiers are characteristic features of MM. In the past decade, several studies have suggested epigenetic mechanisms via DNA methylation, histone modifications and non-coding RNAs as important contributing factors in MM with impacts on disease initiation, progression, clonal heterogeneity and response to treatment. Herein we review the present view and knowledge that has accumulated over the past decades on the role of epigenetics in MM, with focus on the interplay between epigenetic mechanisms and the potential use of epigenetic inhibitors as future treatment modalities for MM.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Animals , Humans , Tumor Microenvironment
5.
Epigenomes ; 2(3)2018 Sep.
Article in English | MEDLINE | ID: mdl-30761216

ABSTRACT

The enhancer of zeste homolog 2 (EZH2) is the enzymatic subunit of the polycomb repressive complex 2 (PRC2) that exerts important functions during normal development as well as disease. PRC2 through EZH2 tri-methylates histone H3 lysine tail residue 27 (H3K27me3), a modification associated with repression of gene expression programs related to stem cell self-renewal, cell cycle, cell differentiation, and cellular transformation. EZH2 is deregulated and subjected to gain of function or loss of function mutations, and hence functions as an oncogene or tumor suppressor gene in a context-dependent manner. The development of highly selective inhibitors against the histone methyltransferase activity of EZH2 has also contributed to insight into the role of EZH2 and PRC2 in tumorigenesis, and their potential as therapeutic targets in cancer. EZH2 can function as an oncogene in multiple myeloma (MM) by repressing tumor suppressor genes that control apoptosis, cell cycle control and adhesion properties. Taken together these findings have raised the possibility that EZH2 inhibitors could be a useful therapeutic modality in MM alone or in combination with other targeted agents in MM. Therefore, we review the current knowledge on the regulation of EZH2 and its biological impact in MM, the anti-myeloma activity of EZH2 inhibitors and their potential as a targeted therapy in MM.

6.
Oncotarget ; 8(61): 103731-103743, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29262596

ABSTRACT

Multiple myeloma (MM) is a tumor of plasmablasts/plasma cells (PCs) characterized by the expansion of malignant PCs with complex genetic aberrations in the bone marrow (BM). Recent reports, by us and others, have highlighted the polycomb group (PcG) proteins as potential targets for therapy in MM. The PcG protein BMI-1 of the polycomb repressive complex 1 (PRC1) has been reported to be overexpressed and to possess oncogenic functions in MM. Herein, we report on the anti-myeloma effects of the BMI-1 inhibitor PTC-209 and demonstrate that PTC-209 is a potent anti-myeloma agent in vitro using MM cell lines and primary MM cells. We show that PTC-209 reduces the viability of MM cells via induction of apoptosis and reveal that the anti-MM actions of PTC-209 are mediated by on-target effects i.e. downregulation of BMI-1 protein and the associated repressive histone mark H2AK119ub, leaving other PRC1 subunits such as CBX-7 and the catalytic subunit RING1B unaffected. Importantly, we demonstrate that PTC-209 exhibits synergistic and additive anti-myeloma activity when combined with other epigenetic inhibitors targeting EZH2 and BET bromodomains. Collectively, these data qualify BMI-1 as a candidate for targeted therapy in MM alone or in combinations with epigenetic inhibitors directed to PRC2/EZH2 or BET bromodomains.

7.
RNA Dis ; 4(2)2017.
Article in English | MEDLINE | ID: mdl-28664185

ABSTRACT

We have previously presented the histone methyltransferase enhancer of zeste homolog 2 (EZH2) of the polycomb repressive complex 2 (PRC2) as a potential therapeutic target in Multiple Myeloma (MM). In a recent article in Oncotarget by Alzrigat. et al. 2017, we have reported on the novel finding that EZH2 inhibition using the highly selective inhibitor of EZH2 enzymatic activity, UNC1999, reactivated the expression of microRNA genes previously reported to be underexpressed in MM. Among these, we have identified miR-125a-3p and miR-320c as potential tumor suppressor microRNAs as they were predicted to target MM-associated oncogenes; IRF-4, XBP-1 and BLIMP-1. We also found EZH2 inhibition to reactivate the expression of miR-494, a previously reported regulator of the c-MYC oncogene. In addition, we could report that EZH2 inhibition downregulated the expression of a few well described oncogenic microRNAs in MM. The data from our recent article are here highlighted as it shed a new light onto the oncogenic function of the PRC2 in MM. These data further strengthen the notion that the PRC2 complex may be of potential therapeutic interest.

8.
J Cell Physiol ; 232(10): 2722-2730, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28112398

ABSTRACT

Increasing number of studies have shown nuclear localization of the insulin-like growth factor 1 receptor (nIGF-1R) in tumor cells and its links to adverse clinical outcome in various cancers. Any obvious cell physiological roles of nIGF-1R have, however, still not been disclosed. Previously, we reported that IGF-1R translocates to cell nucleus and modulates gene expression by binding to enhancers, provided that the receptor is SUMOylated. In this study, we constructed stable transfectants of wild type IGF1R (WT) and triple-SUMO-site-mutated IGF1R (TSM) using igf1r knockout mouse fibroblasts (R-). Cell clones (R-WT and R-TSM) expressing equal amounts of IGF-1R were selected for experiments. Phosphorylation of IGF-1R, Akt, and Erk upon IGF-1 stimulation was equal in R-WT and R-TSM. WT was confirmed to enter nuclei. TSM did also undergo nuclear translocation, although to a lesser extent. This may be explained by that TSM heterodimerizes with insulin receptor, which is known to translocate to cell nuclei. R-WT proliferated substantially faster than R-TSM, which did not differ significantly from the empty vector control. Upon IGF-1 stimulation G1-S-phase progression of R-WT increased from 12 to 38%, compared to 13 to 20% of R-TSM. The G1-S progression of R-WT correlated with increased expression of cyclin D1, A, and CDK2, as well as downregulation of p27. This suggests that SUMO-IGF-1R affects upstream mechanisms that control and coordinate expression of cell cycle regulators. Further studies to identify such SUMO-IGF-1R dependent mechanisms seem important.


Subject(s)
Cell Proliferation , Fibroblasts/metabolism , G1 Phase , Receptor, IGF Type 1/metabolism , Receptors, Somatomedin/metabolism , S Phase , Sumoylation , Animals , Cells, Cultured , Cyclin A/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Genotype , Mice, Knockout , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/deficiency , Receptor, IGF Type 1/genetics , Receptors, Somatomedin/deficiency , Receptors, Somatomedin/genetics , Signal Transduction , Time Factors , Transfection
9.
Oncotarget ; 8(6): 10213-10224, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28052011

ABSTRACT

Multiple Myeloma (MM) is a plasma cell tumor localized to the bone marrow (BM). Despite the fact that current treatment strategies have improved patients' median survival time, MM remains incurable. Epigenetic aberrations are emerging as important players in tumorigenesis making them attractive targets for therapy in cancer including MM. Recently, we suggested the polycomb repressive complex 2 (PRC2) as a common denominator of gene silencing in MM and presented the PRC2 enzymatic subunit enhancer of zeste homolog 2 (EZH2) as a potential therapeutic target in MM. Here we further dissect the anti-myeloma mechanisms mediated by EZH2 inhibition and show that pharmacological inhibition of EZH2 reduces the expression of MM-associated oncogenes; IRF-4, XBP-1, PRDM1/BLIMP-1 and c-MYC. We show that EZH2 inhibition reactivates the expression of microRNAs with tumor suppressor functions predicted to target MM-associated oncogenes; primarily miR-125a-3p and miR-320c. ChIP analysis reveals that miR-125a-3p and miR-320c are targets of EZH2 and H3K27me3 in MM cell lines and primary cells. Our results further highlight that polycomb-mediated silencing in MM includes microRNAs with tumor suppressor activity. This novel role strengthens the oncogenic features of EZH2 and its potential as a therapeutic target in MM.


Subject(s)
Antineoplastic Agents/pharmacology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Genes, Tumor Suppressor , MicroRNAs/genetics , Multiple Myeloma/drug therapy , Oncogenes , Pyridones/pharmacology , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , MicroRNAs/metabolism , Multiple Myeloma/enzymology , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/drug effects , Transcriptome , Tumor Cells, Cultured , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
10.
Hum Mutat ; 37(9): 964-75, 2016 09.
Article in English | MEDLINE | ID: mdl-27328812

ABSTRACT

Chromatin-remodeling factors are required for a wide range of cellular and biological processes including development and cognition, mainly by regulating gene expression. As these functions would predict, deregulation of chromatin-remodeling factors causes various disease syndromes, including neurodevelopmental disorders. Recent reports have linked mutations in several genes coding for chromatin-remodeling factors to intellectual disability (ID). Here, we used exome sequencing and identified a nonsynonymous de novo mutation in BAZ1A (NM_182648.2:c.4043T > G, p.Phe1348Cys), encoding the ATP-utilizing chromatin assembly and remodeling factor 1 (ACF1), in a patient with unexplained ID. ACF1 has been previously reported to bind to the promoter of the vitamin D receptor (VDR)-regulated genes and suppress their expression. Our results show that the patient displays decreased binding of ACF1 to the promoter of the VDR-regulated gene CYP24A1. Using RNA sequencing, we find that the mutation affects the expression of genes involved in several pathways including vitamin D metabolism, Wnt signaling and synaptic formation. RNA sequencing of BAZ1A knockdown cells and Baz1a knockout mice revealed that BAZ1A carry out distinctive functions in different tissues. We also demonstrate that BAZ1A depletion influence the expression of genes important for nervous system development and function. Our data point to an important role for BAZ1A in neurodevelopment, and highlight a possible link for BAZ1A to ID.


Subject(s)
Intellectual Disability/genetics , Nervous System/growth & development , Transcription Factors/genetics , Transcription Factors/metabolism , Amino Acid Substitution , Animals , Cell Line , Chromosomal Proteins, Non-Histone , Exome , Gene Regulatory Networks , Humans , Mice , Promoter Regions, Genetic , Receptors, Calcitriol/metabolism , Sequence Analysis, DNA , Sequence Analysis, RNA , Synaptic Potentials , Tissue Distribution , Wnt Signaling Pathway
11.
Oncotarget ; 7(6): 6809-23, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26755663

ABSTRACT

Multiple myeloma (MM) is a malignancy of the antibody-producing plasma cells. MM is a highly heterogeneous disease, which has hampered the identification of a common underlying mechanism for disease establishment as well as the development of targeted therapy. Here we present the first genome-wide profiling of histone H3 lysine 27 and lysine 4 trimethylation in MM patient samples, defining a common set of active H3K4me3-enriched genes and silent genes marked by H3K27me3 (H3K27me3 alone or bivalent) unique to primary MM cells, when compared to normal bone marrow plasma cells. Using this epigenome profile, we found increased silencing of H3K27me3 targets in MM patients at advanced stages of the disease, and the expression pattern of H3K27me3-marked genes correlated with poor patient survival. We also demonstrated that pharmacological inhibition of EZH2 had anti-myeloma effects in both MM cell lines and CD138+ MM patient cells. In addition, EZH2 inhibition decreased the global H3K27 methylation and induced apoptosis. Taken together, these data suggest an important role for the Polycomb repressive complex 2 (PRC2) in MM, and highlights the PRC2 component EZH2 as a potential therapeutic target in MM.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Histones/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Polycomb-Group Proteins/genetics , Chromatin/metabolism , Down-Regulation , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Profiling , Histones/genetics , Humans , Lysine/metabolism , Methylation , Molecular Targeted Therapy , Multiple Myeloma/metabolism , Polycomb-Group Proteins/metabolism
12.
Cell Cycle ; 15(12): 1558-71, 2016 06 17.
Article in English | MEDLINE | ID: mdl-25483050

ABSTRACT

CGGBP1 (CGG triplet repeat-binding protein 1) regulates cell proliferation, stress response, cytokinesis, telomeric integrity and transcription. It could affect these processes by modulating target gene expression under different conditions. Identification of CGGBP1-target genes and their regulation could reveal how a transcription regulator affects such diverse cellular processes. Here we describe the mechanisms of differential gene expression regulation by CGGBP1 in quiescent or growing cells. By studying global gene expression patterns and genome-wide DNA-binding patterns of CGGBP1, we show that a possible mechanism through which it affects the expression of RNA Pol II-transcribed genes in trans depends on Alu RNA. We also show that it regulates Alu transcription in cis by binding to Alu promoter. Our results also indicate that potential phosphorylation of CGGBP1 upon growth stimulation facilitates its nuclear retention, Alu-binding and dislodging of RNA Pol III therefrom. These findings provide insights into how Alu transcription is regulated in response to growth signals.


Subject(s)
Alu Elements , DNA-Binding Proteins/genetics , Fibroblasts/metabolism , RNA Polymerase II/genetics , Transcription, Genetic , Base Sequence , Biological Factors/pharmacology , DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Profiling , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Lentivirus/genetics , Lentivirus/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , RNA Polymerase II/metabolism , Sequence Alignment , Serum/chemistry
13.
Oncotarget ; 6(24): 20621-35, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26036313

ABSTRACT

Despite the introduction of new treatment options for multiple myeloma (MM), a majority of patients relapse due to the development of resistance. Unraveling new mechanisms underlying resistance could lead to identification of possible targets for combinatorial treatment. Using TRAF3 deleted/mutated MM cell lines, we evaluated the role of the cellular inhibitor of apoptosis 2 (cIAP2) in drug resistance and uncovered the plausible mechanisms underlying this resistance and possible strategies to overcome this by combinatorial treatment. In MM, cIAP2 is part of the gene signature of aberrant NF-κB signaling and is heterogeneously expressed amongst MM patients. In cIAP2 overexpressing cells a decreased sensitivity to the proteasome inhibitors bortezomib, MG132 and carfilzomib was observed. Gene expression analysis revealed that 440 genes were differentially expressed due to cIAP2 overexpression. Importantly, the data imply that cIAPs are rational targets for combinatorial treatment in the population of MM with deleted/mutated TRAF3. Indeed, we found that treatment with the IAP inhibitor AT-406 enhanced the anti-MM effect of bortezomib in the investigated cell lines. Taken together, our results show that cIAP2 is an important factor mediating bortezomib resistance in MM cells harboring TRAF3 deletion/mutation and therefore should be considered as a target for combinatorial treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Proteasome Inhibitors/pharmacology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/metabolism , Apoptosis/drug effects , Apoptosis/physiology , Azocines/administration & dosage , Azocines/pharmacology , Baculoviral IAP Repeat-Containing 3 Protein , Benzhydryl Compounds/administration & dosage , Benzhydryl Compounds/pharmacology , Bortezomib/administration & dosage , Bortezomib/pharmacology , Case-Control Studies , Cell Line, Tumor , Drug Resistance, Neoplasm , Drug Synergism , HEK293 Cells , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/genetics , Multiple Myeloma/pathology , NF-kappa B/metabolism , TNF Receptor-Associated Factor 3/deficiency , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
14.
J Exp Med ; 212(6): 833-43, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25987724

ABSTRACT

NF-κB is constitutively activated in chronic lymphocytic leukemia (CLL); however, the implicated molecular mechanisms remain largely unknown. Thus, we performed targeted deep sequencing of 18 core complex genes within the NF-κB pathway in a discovery and validation CLL cohort totaling 315 cases. The most frequently mutated gene was NFKBIE (21/315 cases; 7%), which encodes IκBε, a negative regulator of NF-κB in normal B cells. Strikingly, 13 of these cases carried an identical 4-bp frameshift deletion, resulting in a truncated protein. Screening of an additional 377 CLL cases revealed that NFKBIE aberrations predominated in poor-prognostic patients and were associated with inferior outcome. Minor subclones and/or clonal evolution were also observed, thus potentially linking this recurrent event to disease progression. Compared with wild-type patients, NFKBIE-deleted cases showed reduced IκBε protein levels and decreased p65 inhibition, along with increased phosphorylation and nuclear translocation of p65. Considering the central role of B cell receptor (BcR) signaling in CLL pathobiology, it is notable that IκBε loss was enriched in aggressive cases with distinctive stereotyped BcR, likely contributing to their poor prognosis, and leading to an altered response to BcR inhibitors. Because NFKBIE deletions were observed in several other B cell lymphomas, our findings suggest a novel common mechanism of NF-κB deregulation during lymphomagenesis.


Subject(s)
Gene Expression Regulation, Leukemic , I-kappa B Kinase/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , NF-kappa B/metabolism , Cell Nucleus/metabolism , Cell Survival , Chromosome Aberrations , Cohort Studies , Cytoplasm/metabolism , DNA Mutational Analysis , Frameshift Mutation , Gene Deletion , Gene Expression Profiling , Humans , I-kappa B Kinase/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell, Marginal Zone/metabolism , Lymphoma, Mantle-Cell/metabolism , Oligonucleotide Array Sequence Analysis , Receptors, Antigen, B-Cell/metabolism , Signal Transduction , Treatment Outcome
15.
Oncotarget ; 5(22): 11193-208, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25008202

ABSTRACT

The ABT-analogous 737, 263 and 199 are BH3 mimetics showing potent anti-myeloma (MM) activity, but only on defined molecular subgroups of MM patients presenting a Bcl-2high/Mcl-1low profile. IGF-1 is a major survival factor in MM regulating the expression of Bcl-2 proteins and might therefore be a resistance factor to these ABT-analogous. We first show that IGF-1 protected human MM cell lines (HMCLs) against ABT-737. Concurrently, the IGF-1 receptor inhibitor picropodophyllin (PPP) synergistically sensitized HMCL, primary human MM and murine 5T33MM cells to ABT-737 and ABT-199 by further decreasing cell viability and enhancing apoptosis. Knockdown of Bcl-2 by shRNA protected MM cells to ABT-737, while Mcl-1 shRNA sensitized the cells. PPP overcame the Bcl-2 dependency of ABT-737, but failed to completely overcome the protective effect of Mcl-1. In vivo, co-treatment of 5T33MM bearing mice significantly decreased tumor burden and prolonged overall survival both in a prophylactic and therapeutic setting. Interestingly, proteasome inhibitor resistant CD138- 5T33MM cells were more sensitive to ABT-737, whereas PPP alone targeted the CD138+ cells more effectively. After co-treatment, both subpopulations were targeted equally. Together, the combination of an IGF-1R inhibitor and an ABT-analogue displays synergistic anti-myeloma activity providing the rational for further (pre)clinical testing.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biphenyl Compounds/pharmacology , Multiple Myeloma/drug therapy , Nitrophenols/pharmacology , Podophyllotoxin/analogs & derivatives , Receptor, IGF Type 1/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Biomimetic Materials/administration & dosage , Biomimetic Materials/pharmacology , Biphenyl Compounds/administration & dosage , Cell Line, Tumor , Drug Synergism , Humans , Mice , Mice, Inbred C57BL , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Nitrophenols/administration & dosage , Piperazines/administration & dosage , Piperazines/pharmacology , Podophyllotoxin/administration & dosage , Podophyllotoxin/pharmacology , Sulfonamides/administration & dosage
16.
Cancer Res ; 72(20): 5348-62, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22952216

ABSTRACT

Multiple myeloma (MM) is a B-cell malignancy characterized by the expansion of clonal plasma blasts/plasma cells within the bone marrow that relies on multiple signaling cascades, including tyrosine kinase activated pathways, to proliferate and evade cell death. Despite emerging new treatment strategies, multiple myeloma remains at present incurable. Thus, novel approaches targeting several signaling cascades by using the multi-tyrosine kinase inhibitor (TKI), sorafenib, seem a promising treatment approach for multiple myeloma. Here, we show that sorafenib induces cell death in multiple myeloma cell lines and in CD138(+)-enriched primary multiple myeloma patient samples in a caspase-dependent and -independent manner. Furthermore, sorafenib has a strong antitumoral and -angiogenic activity in the 5T33MM mouse model leading to increased overall survival. Multiple myeloma cells undergo autophagy in response to sorafenib, and inhibition of this cytoprotective pathway potentiated the efficacy of this TKI. Mcl-1, a survival factor in multiple myeloma, is downregulated at the protein level by sorafenib allowing for the execution of cell death, as ectopic overexpression of this protein protects multiple myeloma cells. Concomitant targeting of Mcl-1 by sorafenib and of Bcl-2/Bcl-xL by the antagonist ABT737 improves the efficacy of sorafenib in multiple myeloma cell lines and CD138(+)-enriched primary cells in the presence of bone marrow stromal cells. Altogether, our data support the use of sorafenib as a novel therapeutic modality against human multiple myeloma, and its efficacy may be potentiated in combination with ABT737.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzenesulfonates/therapeutic use , Multiple Myeloma/drug therapy , Pyridines/therapeutic use , Adult , Aged , Aged, 80 and over , Animals , Base Sequence , DNA Primers , Disease Models, Animal , Female , Flow Cytometry , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Multiple Myeloma/pathology , Niacinamide/analogs & derivatives , Phenylurea Compounds , Sorafenib
17.
Am J Hematol ; 87(4): 361-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22374828

ABSTRACT

Mantle cell lymphoma (MCL) and chronic lymphocytic leukemia (CLL) are mature CD5(+) B-cell malignancies with different biological/clinical characteristics. We recently reported an association between different prognostic subgroups of CLL (i.e., IGHV mutated and unmutated) and genomic methylation pattern. However, the relationship between DNA methylation and prognostic markers, such as the proliferation gene expression signature, has not been investigated in MCL. We applied high-resolution methylation microarrays (27,578 CpG sites) to assess the global DNA methylation profiles in 20 MCL (10 each with high/low proliferation signature) and 30 CLL (15 poor-prognostic IGHV unmutated subset #1 and 15 good-prognostic IGHV mutated subset #4) samples. Notably, MCL and each CLL subset displayed distinct genomic methylation profiles. After unsupervised hierarchical clustering, 17/20 MCL cases formed a cluster separate from CLL, while CLL subsets #1 and #4 formed subclusters. Surprisingly, few differentially methylated genes (n = 6) were identified between high vs. low proliferation MCL. In contrast, distinct methylation profiles were demonstrated for MCL and CLL. Importantly, certain functional classes of genes were preferentially methylated in either disease. For instance, developmental genes, in particular homeobox transcription factor genes (e.g., HLXB9, HOXA13), were more highly methylated in MCL, whereas apoptosis-related genes were enriched among targets methylated in CLL (e.g., CYFIP2, NR4A1). Results were validated using pyrosequencing, RQ-PCR and reexpression of specific genes. In summary, the methylation profile of MCL was homogeneous and no correlation with the proliferation signature was observed. Compared to CLL, however, marked differences were discovered such as the preferential methylation of homeobox genes in MCL.


Subject(s)
DNA Methylation , DNA, Neoplasm/chemistry , Gene Expression Regulation, Neoplastic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Lymphoma, Mantle-Cell/genetics , Apoptosis Regulatory Proteins/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Division , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , CpG Islands/drug effects , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA Methylation/drug effects , DNA, Neoplasm/genetics , Decitabine , Female , Gene Expression Regulation, Leukemic , Gene Expression Regulation, Neoplastic/drug effects , Genes, Homeobox , Genes, Immunoglobulin , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Immunoglobulin Heavy Chains/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphoma, Mantle-Cell/metabolism , Male , Neoplasm Proteins/genetics , Sequence Analysis, DNA , Transcription Factors/genetics
18.
Clin Cancer Res ; 18(8): 2230-9, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22392915

ABSTRACT

PURPOSE: We have previously shown the use of the insulin-like growth factor type 1 receptor tyrosine kinase (IGF-1RTK) inhibitor picropodophyllin (PPP) as an attractive strategy to combat multiple myeloma (MM) in vitro and in vivo. After a combinatorial drug screening, the histone deacetylase inhibitor LBH589 was shown to act in synergy with PPP reducing survival of MM cells. In this study, we tried to elucidate the molecular mechanisms underlying this combinatorial effect. EXPERIMENTAL DESIGN: The in vitro anti-MM effects of PPP and LBH589 alone and in combination were evaluated by studying apoptosis, cell cycle distribution, and downstream transcriptome using both human MM cell lines and cells from the murine 5T3MM model. In vivo the effect on survival of 5T33MM-inoculated mice was evaluated. RESULTS: In the human MM cell line RPMI8226, treatment with PPP and LBH589 in combination resulted in a five-fold increase of apoptosis, and an additive effect on the cleavage of the active forms of caspase-8 was observed as compared with the single drug treatments. Cell cycle analysis revealed an accumulation of cells in the G(2)-M phase and subsequent downregulation of cell cycle regulating proteins. These data were also confirmed in the 5T33MM cells in vitro. Also, the transcriptome was analyzed by Affymetrix arrays showing gene expression alterations mainly in categories of genes regulating apoptosis and cell adhesion. Combined treatment in vivo resulted in a significantly prolonged survival of 5T33MM-inoculated mice. CONCLUSIONS: The results indicate an improved MM treatment opportunity in using a combination of PPP and LBH589.


Subject(s)
Hydroxamic Acids/therapeutic use , Multiple Myeloma/drug therapy , Podophyllotoxin/analogs & derivatives , Animals , Apoptosis/drug effects , Caspase 8/metabolism , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Cycle Proteins/biosynthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , G2 Phase Cell Cycle Checkpoints , Gene Expression Profiling , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/pharmacology , Indoles , Mice , Mice, Inbred C57BL , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Panobinostat , Podophyllotoxin/pharmacology , Podophyllotoxin/therapeutic use , Receptor, IGF Type 1/antagonists & inhibitors , Transcriptome/drug effects , Tumor Cells, Cultured
19.
Ups J Med Sci ; 117(2): 166-77, 2012 May.
Article in English | MEDLINE | ID: mdl-22348393

ABSTRACT

Multiple myeloma (MM) is a B cell malignancy characterized by the expansion of clonal plasmablast/plasma cells within the bone-marrow. It is well established that the bone-marrow microenvironment has a pivotal role in providing critical cytokines and cell-cell interactions to support the growth and survival of the MM tumor clone. The pathogenesis of MM is, however, only fragmentarily understood. Detailed genomic analysis reveals a heterogeneous and complex pattern of structural and numerical chromosomal aberrations. In this review we will discuss some of the recent results on the functional role and potential clinical use of the IGF-1R, one of the major mediators of growth and survival for MM. We will also describe some of our results on epigenetic gene silencing in MM, as it may indeed constitute a novel basis for the understanding of tumor initiation and maintenance in MM and thus may change the current view on treatment strategies for MM.


Subject(s)
Epigenesis, Genetic , Gene Silencing , Multiple Myeloma/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Multiple Myeloma/genetics
20.
PLoS One ; 6(3): e14757, 2011 Mar 14.
Article in English | MEDLINE | ID: mdl-21423728

ABSTRACT

BACKGROUND: Picropodophyllin (PPP) is a promising novel anti-neoplastic agent that efficiently kills tumor cells in vitro and causes tumor regression and increased survival in vivo. We have previously reported that PPP treatment induced moderate tolerance in two out of 10 cell lines only, and here report the acquired genomic and expression alterations associated with PPP selection over 1.5 years of treatment. METHODOLOGY/PRINCIPAL FINDINGS: Copy number alterations monitored using metaphase and array-based comparative genomic hybridization analyses revealed largely overlapping alterations in parental and maximally tolerant cells. Gain/amplification of the MYC and PVT1 loci in 8q24.21 were verified on the chromosome level. Abnormalities observed in connection to PPP treatment included regular gains and losses, as well as homozygous losses in 10q24.1-q24.2 and 12p12.3-p13.2 in one of the lines and amplification at 5q11.2 in the other. Abnormalities observed in both tolerant derivatives include amplification/gain of 5q11.2, gain of 11q12.1-q14.3 and gain of 13q33.3-qter. Using Nexus software analysis we combined the array-CGH data with data from gene expression profilings and identified genes that were altered in both inputs. A subset of genes identified as downregulated (ALDH1A3, ANXA1, TLR4 and RAB5A) or upregulated (COX6A1, NFIX, ME1, MAPK and TAP2) were validated by siRNA in the tolerant or parental cells to alter sensitivity to PPP and confirmed to alter sensitivity to PPP in further cell lines. CONCLUSIONS: Long-term PPP selection lead to altered gene expression in PPP tolerant cells with increase as well as decrease of genes involved in cell death such as PTEN and BCL2. In addition, acquired genomic copy number alterations were observed that were often reflected by altered mRNA expression levels for genes in the same regions.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Neoplasms/genetics , Podophyllotoxin/analogs & derivatives , Allelic Imbalance/drug effects , Allelic Imbalance/genetics , Cell Line, Tumor , Chromosome Aberrations/drug effects , Cluster Analysis , Comparative Genomic Hybridization , DNA Copy Number Variations/genetics , DNA Methylation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genotype , Humans , In Situ Hybridization, Fluorescence , Metaphase/drug effects , Podophyllotoxin/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/metabolism , Reproducibility of Results , Spectral Karyotyping
SELECTION OF CITATIONS
SEARCH DETAIL
...