Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Haematologica ; 109(2): 567-577, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37496441

ABSTRACT

Multiple myeloma is a heterogeneous hematological disease that originates from the bone marrow and is characterized by the monoclonal expansion of malignant plasma cells. Despite novel therapies, multiple myeloma remains clinically challenging. A common feature among patients with poor prognosis is the increased activity of the epigenetic silencer EZH2, which is the catalytic subunit of the PRC2. Interestingly, the recruitment of PRC2 lacks sequence specificity and, to date, the molecular mechanisms that define which genomic locations are destined for PRC2-mediated silencing remain unknown. The presence of a long non-coding RNA (lncRNA)-binding pocket on EZH2 suggests that lncRNA could potentially mediate PRC2 recruitment to specific genomic regions. Here, we coupled RNA immunoprecipitation sequencing, RNA-sequencing and chromatin immunoprecipitation-sequencing analysis of human multiple myeloma primary cells and cell lines to identify potential lncRNA partners to EZH2. We found that the lncRNA plasmacytoma variant translocation 1 (PVT1) directly interacts with EZH2 and is overexpressed in patients with a poor prognosis. Moreover, genes predicted to be targets of PVT1 exhibited H3K27me3 enrichment and were associated with pro-apoptotic and tumor suppressor functions. In fact, PVT1 inhibition independently promotes the expression of the PRC2 target genes ZBTB7C, RNF144A and CCDC136. Altogether, our work suggests that PVT1 is an interacting partner in PRC2-mediated silencing of tumor suppressor and pro-apoptotic genes in multiple myeloma, making it a highly interesting potential therapeutic target.


Subject(s)
Multiple Myeloma , RNA, Long Noncoding , Humans , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Multiple Myeloma/drug therapy , Enhancer of Zeste Homolog 2 Protein/genetics , RNA, Long Noncoding/genetics , Cell Line, Tumor , Genomics , Intracellular Signaling Peptides and Proteins
2.
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
3.
Front Oncol ; 13: 1303677, 2023.
Article in English | MEDLINE | ID: mdl-38148842

ABSTRACT

Extensive genome-wide sequencing efforts have unveiled the intricate regulatory potential of long non-protein coding RNAs (lncRNAs) within the domain of haematological malignancies. Notably, lncRNAs have been found to directly modulate chromatin architecture, thereby impacting gene expression and disease progression by interacting with DNA, RNA, and proteins in a tissue- or condition-specific manner. Furthermore, recent studies have highlighted the intricate epigenetic control of lncRNAs in cancer. Consequently, this provides a rationale to explore the possibility of therapeutically targeting lncRNAs themselves or the epigenetic mechanisms that govern their activity. Within the scope of this review, we will assess the current state of knowledge regarding the epigenetic regulation of lncRNAs and how, in turn, lncRNAs contribute to chromatin remodelling in the context of multiple myeloma.

4.
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
5.
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.

6.
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
7.
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.

8.
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
9.
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
10.
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
11.
Biochem Pharmacol ; 87(2): 284-91, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24200998

ABSTRACT

AKN-028 is a novel tyrosine kinase inhibitor with preclinical activity in acute myeloid leukemia (AML), presently undergoing investigation in a phase I/II study. It is a potent inhibitor of the FMS-like kinase 3 (FLT3) but shows in vitro activity in a wide range of AML samples. In the present study, we have characterized the effects of AKN-028 on AML cells in more detail. AKN-028 induced a dose-dependent G0/1 arrest in AML cell line MV4-11. Treatment with AKN-028 caused significantly altered gene expression in all AML cell types tested (430 downregulated, 280 upregulated transcripts). Subsequent gene set enrichment analysis revealed enrichment of genes associated with the proto-oncogene and cell cycle regulator c-Myc among the downregulated genes in both AKN-028 and midostaurin treated cells. Kinase activity profiling in AML cell lines and primary AML samples showed that tyrosine kinase activity, but not serine/threonine kinase activity, was inhibited by AKN-028 in a dose dependent manner in all samples tested, reaching approximately the same level of kinase activity. Cells sensitive to AKN-028 showed a higher overall tyrosine kinase activity than more resistant ones, whereas serine/threonine kinase activity was similar for all primary AML samples. In summary, AKN-028 induces cell cycle arrest in AML cells, downregulates Myc-associated genes and affect several signaling pathways. AML cells with high global tyrosine kinase activity seem to be more sensitive to the cytotoxic effect of AKN-028 in vitro.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Checkpoints/drug effects , Down-Regulation/drug effects , Genes, myc/drug effects , Indoles/pharmacology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazines/pharmacology , Antineoplastic Agents/therapeutic use , Cell Cycle Checkpoints/genetics , Dose-Response Relationship, Drug , Down-Regulation/genetics , Genes, myc/genetics , HL-60 Cells , Humans , Indoles/therapeutic use , Leukemia, Myeloid, Acute/enzymology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Mas , Pyrazines/therapeutic use , Tumor Cells, Cultured
12.
PLoS One ; 5(7): e11483, 2010 Jul 09.
Article in English | MEDLINE | ID: mdl-20634887

ABSTRACT

Multiple myeloma (MM) is a genetically heterogeneous disease, which to date remains fatal. Finding a common mechanism for initiation and progression of MM continues to be challenging. By means of integrative genomics, we identified an underexpressed gene signature in MM patient cells compared to normal counterpart plasma cells. This profile was enriched for previously defined H3K27-tri-methylated genes, targets of the Polycomb group (PcG) proteins in human embryonic fibroblasts. Additionally, the silenced gene signature was more pronounced in ISS stage III MM compared to stage I and II. Using chromatin immunoprecipitation (ChIP) assay on purified CD138+ cells from four MM patients and on two MM cell lines, we found enrichment of H3K27me3 at genes selected from the profile. As the data implied that the Polycomb-targeted gene profile would be highly relevant for pharmacological treatment of MM, we used two compounds to chemically revert the H3K27-tri-methylation mediated gene silencing. The S-adenosylhomocysteine hydrolase inhibitor 3-Deazaneplanocin (DZNep) and the histone deacetylase inhibitor LBH589 (Panobinostat), reactivated the expression of genes repressed by H3K27me3, depleted cells from the PRC2 component EZH2 and induced apoptosis in human MM cell lines. In the immunocompetent 5T33MM in vivo model for MM, treatment with LBH589 resulted in gene upregulation, reduced tumor load and increased overall survival. Taken together, our results reveal a common gene signature in MM, mediated by gene silencing via the Polycomb repressor complex. The importance of the underexpressed gene profile in MM tumor initiation and progression should be subjected to further studies.


Subject(s)
Multiple Myeloma/drug therapy , Repressor Proteins/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/therapeutic use , Adenosylhomocysteinase/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/genetics , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cells, Cultured , Chemokine CXCL12/genetics , Chemokine CXCL12/metabolism , Chromatin Immunoprecipitation , Computational Biology , DNA Methylation/drug effects , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Indoles , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Kaplan-Meier Estimate , Mice , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Panobinostat , Polycomb-Group Proteins , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Mol Med Rep ; 1(2): 263-8, 2008.
Article in English | MEDLINE | ID: mdl-21479407

ABSTRACT

The SMARCA3 gene was recently found to be a common target for methylation in colon and gastric cancer, suggesting it has possible tumor suppressor activity. To determine whether SMARCA3 plays a role in colorectal and/or gastric cancer predisposition, a mutation screening of the gene was performed in affected index cases from 20 Swedish families with colorectal and/or gastric cancer. Notably, one family included in the screening exhibited suggestive linkage to the region on chromosome 3q that harbors the SMARCA3 gene. In addition to known polymorphisms, nine novel variants - none of them clearly pathogenic - were detected. Seven of these variants were further tested in an additional 287 patients with a family history of the disease, but their frequency was found to not be significantly different from that observed in the controls. In conclusion, although a very low effect of some variants could not be excluded, it seems that the SMARCA3 gene does not play an important role in the predisposition to colorectal and gastric cancer.

SELECTION OF CITATIONS
SEARCH DETAIL