Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(1): 113644, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38180837

ABSTRACT

Extensive remodeling of the female mammary epithelium during development and pregnancy has been linked to cancer susceptibility. The faithful response of mammary epithelial cells (MECs) to hormone signaling is key to avoiding breast cancer development. Here, we show that lactogenic differentiation of murine MECs requires silencing of genes encoding ribosomal RNA (rRNA) by the antisense transcript PAPAS. Accordingly, knockdown of PAPAS derepresses rRNA genes, attenuates the response to lactogenic hormones, and induces malignant transformation. Restoring PAPAS levels in breast cancer cells reduces tumorigenicity and lung invasion and activates many interferon-regulated genes previously linked to metastasis suppression. Mechanistically, PAPAS transcription depends on R-loop formation at the 3' end of rRNA genes, which is repressed by RNase H1 and replication protein A (RPA) overexpression in breast cancer cells. Depletion of PAPAS and upregulation of RNase H1 and RPA in human breast cancer underpin the clinical relevance of our findings.


Subject(s)
Breast Neoplasms , Mammary Glands, Animal , Pregnancy , Female , Mice , Animals , Humans , Mammary Glands, Animal/metabolism , Breast/metabolism , Cell Differentiation , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism
2.
Int J Mol Sci ; 24(6)2023 Mar 11.
Article in English | MEDLINE | ID: mdl-36982453

ABSTRACT

Immortalized cell lines are widely used in vitro tools in oncology and hematology research. While these cell lines represent artificial systems and may accumulate genetic aberrations with each passage, they are still considered valuable models for pilot, preliminary, and screening studies. Despite their limitations, cell lines are cost-effective and provide repeatable and comparable results. Choosing the appropriate cell line for acute myeloid leukemia (AML) research is crucial for obtaining reliable and relevant results. Several factors should be considered when selecting a cell line for AML research, such as specific markers and genetic abnormalities associated with different subtypes of AML. It is also essential to evaluate the karyotype and mutational profile of the cell line, as these can influence the behavior and response to the treatment of the cells. In this review, we evaluate immortalized AML cell lines and discuss the issues surrounding them concerning the revised World Health Organization and the French-American-British classifications.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/metabolism , Mutation , Karyotyping , Karyotype
3.
Int J Mol Sci ; 24(4)2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36834787

ABSTRACT

The prognosis for patients with relapsed childhood acute lymphoblastic leukaemia (cALL) remains poor. The main reason for treatment failure is drug resistance, most commonly to glucocorticoids (GCs). The molecular differences between prednisolone-sensitive and -resistant lymphoblasts are not well-studied, thereby precluding the development of novel and targeted therapies. Therefore, the aim of this work was to elucidate at least some aspects of the molecular differences between matched pairs of GC-sensitive and -resistant cell lines. To address this, we carried out an integrated transcriptomic and metabolomic analysis, which revealed that lack of response to prednisolone may be underpinned by alterations in oxidative phosphorylation, glycolysis, amino acid, pyruvate and nucleotide biosynthesis, as well as activation of mTORC1 and MYC signalling, which are also known to control cell metabolism. In an attempt to explore the potential therapeutic effect of inhibiting one of the hits from our analysis, we targeted the glutamine-glutamate-α-ketoglutarate axis by three different strategies, all of which impaired mitochondrial respiration and ATP production and induced apoptosis. Thereby, we report that prednisolone resistance may be accompanied by considerable rewiring of transcriptional and biosynthesis programs. Among other druggable targets that were identified in this study, inhibition of glutamine metabolism presents a potential therapeutic approach in GC-sensitive, but more importantly, in GC-resistant cALL cells. Lastly, these findings may be clinically relevant in the context of relapse-in publicly available datasets, we found gene expression patterns suggesting that in vivo drug resistance is characterised by similar metabolic dysregulation to what we found in our in vitro model.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Prednisolone , Humans , Child , Prednisolone/pharmacology , Glutamine/pharmacology , Drug Resistance, Neoplasm/genetics , Glucocorticoids/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
4.
Br J Haematol ; 198(2): 338-348, 2022 07.
Article in English | MEDLINE | ID: mdl-35468223

ABSTRACT

Aberrant activity of the phosphatidylinositol-3 kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR [PAM]) pathway, as well as suppressed retinoic acid signalling, contribute to enhanced proliferation and the differentiation blockade of immature myeloid cells in acute myeloid leukaemia (AML). Inhibition of the PAM pathway was shown to affect especially mixed-lineage leukaemia-rearranged AML. Here, we sought to test a combined strategy using small molecule inhibitors against members of the PAM signalling pathway in conjunction with all-trans retinoic acid (ATRA) to target a larger group of different AML subtypes. We find that ATRA treatment in combination with inhibition of PI3K (ZSTK474), mTOR (WYE132) or PI3K/mTOR (BEZ235, dactolisib) drastically reduces protein levels of the proto-oncogene MYC. In combination with BEZ235, ATRA treatment led to almost complete eradication of cellular MYC, G1 arrest, loss of clonal capacity and terminal granulocytic differentiation. We demonstrate that PAM inhibitor/ATRA treatment targets MYC via independent mechanisms. While inhibition of the PAM pathway causes MYC phosphorylation at threonine 58 via glycogen synthase kinase 3 beta and subsequent degradation, ATRA reduces its expression. Here, we present an approach using a combination of known drugs to synergistically reduce aberrant MYC levels, thereby effectively blocking proliferation and enabling differentiation in various AML subtypes.


Subject(s)
Leukemia, Myeloid, Acute , Proto-Oncogene Proteins c-akt , Cell Line, Tumor , Cell Proliferation , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases , Tretinoin/pharmacology , Tretinoin/therapeutic use
5.
Cancers (Basel) ; 13(16)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34439223

ABSTRACT

Multiple Myeloma (MM) is a malignancy of plasma cells infiltrating the bone marrow (BM). Many studies have demonstrated the crucial involvement of bone marrow stromal cells in MM progression and drug resistance. Together with the BM microenvironment (BMME), epigenetics also plays a crucial role in MM development. A variety of epigenetic regulators, including histone acetyltransferases (HATs), histone methyltransferases (HMTs) and lysine demethylases (KDMs), are altered in MM, contributing to the disease progression and prognosis. In addition to histone modifications, DNA methylation also plays a crucial role. Among others, aberrant epigenetics involves processes associated with the BMME, like bone homeostasis, ECM remodeling or the development of treatment resistance. In this review, we will highlight the importance of the interplay of MM cells with the BMME in the development of treatment resistance. Additionally, we will focus on the epigenetic aberrations in MM and their role in disease evolution, interaction with the BMME, disease progression and development of drug resistance. We will also briefly touch on the epigenetic treatments currently available or currently under investigation to overcome BMME-driven treatment resistance.

6.
Front Oncol ; 11: 632181, 2021.
Article in English | MEDLINE | ID: mdl-33791218

ABSTRACT

Childhood acute lymphoblastic leukaemia (cALL) accounts for about one third of all paediatric malignancies making it the most common cancer in children. Alterations in tumour cell metabolism were first described nearly a century ago and have been acknowledged as one of the key characteristics of cancers including cALL. Two of the backbone chemotherapeutic agents in the treatment of this disease, Glucocorticoids and L-asparaginase, are exerting their anti-leukaemic effects through targeting cell metabolism. Even though risk stratification and treatment regimens have improved cure rates to nearly 90%, prognosis for relapsed children remains poor. Therefore, new therapeutic approaches are urgently required. Atovaquone is a well-tolerated drug used in the clinic mainly against malaria. Being a ubiquinone analogue, this drug inhibits co-enzyme Q10 of the electron transport chain (ETC) affecting oxidative phosphorylation and cell metabolism. In this study we tested the effect of Atovaquone on cALL cells in vitro. Pharmacologically relevant concentrations of the inhibitor could effectively target mitochondrial respiration in both cALL cell lines (REH and Sup-B15) and primary patient samples. We found that Atovaquone leads to a marked decrease in basal respiration and ATP levels, as well as reduced proliferation, cell cycle arrest, and induction of apoptosis. Importantly, we observed an enhanced anti-leukaemic effect when Atovaquone was combined with the standard chemotherapeutic Idarubicin, or with Prednisolone in an in vitro model of Glucocorticoid resistance. Repurposing of this clinically approved inhibitor renders further investigations, but also presents opportunities for fast-track trials as a single agent or in combination with standard chemotherapeutics.

7.
Cells ; 9(12)2020 12 11.
Article in English | MEDLINE | ID: mdl-33322246

ABSTRACT

The retinoids are a group of compounds including vitamin A and its active metabolite all-trans-retinoic acid (ATRA). Retinoids regulate a variety of physiological functions in multiple organ systems, are essential for normal immune competence, and are involved in the regulation of cell growth and differentiation. Vitamin A derivatives have held promise in cancer treatment and ATRA is used in differentiation therapy of acute promyelocytic leukemia (APL). ATRA and other retinoids have also been successfully applied in a variety of dermatological conditions such as skin cancer, psoriasis, acne, and ichthyosis. Moreover, modulation of retinoic acid receptors and retinoid X (or rexinoid) receptors function may affect dermal cells. The studies using complex genetic models with various combinations of retinoic acid receptors (RARs) and retinoid X (or rexinoid) receptors (RXRs) indicate that retinoic acid and its derivatives have therapeutic potential for a variety of serious dermatological disorders including some malignant conditions. Here, we provide a synopsis of the main advances in understanding the role of ATRA and its receptors in dermatology.


Subject(s)
Skin/drug effects , Tretinoin/pharmacology , Cell Differentiation/drug effects , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/metabolism , Signal Transduction/drug effects , Skin/cytology , Skin/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tretinoin/analogs & derivatives , Tretinoin/metabolism , Tretinoin/therapeutic use
9.
Leukemia ; 33(11): 2628-2639, 2019 11.
Article in English | MEDLINE | ID: mdl-31576004

ABSTRACT

To date, only one subtype of acute myeloid leukemia (AML), acute promyelocytic leukemia (APL) can be effectively treated by differentiation therapy utilizing all-trans retinoic acid (ATRA). Non-APL AMLs are resistant to ATRA. Here we demonstrate that the acetyltransferase GCN5 contributes to ATRA resistance in non-APL AML via aberrant acetylation of histone 3 lysine 9 (H3K9ac) residues maintaining the expression of stemness and leukemia associated genes. We show that inhibition of GCN5 unlocks an ATRA-driven therapeutic response. This response is potentiated by coinhibition of the lysine demethylase LSD1, leading to differentiation in most non-APL AML. Induction of differentiation was not correlated to a specific AML subtype, cytogenetic, or mutational status. Our study shows a previously uncharacterized role of GCN5 in maintaining the immature state of leukemic blasts and identifies GCN5 as a therapeutic target in AML. The high efficacy of the combined epigenetic treatment with GCN5 and LSD1 inhibitors may enable the use of ATRA for differentiation therapy of non-APL AML. Furthermore, it supports a strategy of combined targeting of epigenetic factors to improve treatment, a concept potentially applicable for a broad range of malignancies.


Subject(s)
Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Promyelocytic, Acute/drug therapy , Tretinoin/pharmacology , p300-CBP Transcription Factors/metabolism , Apoptosis , Bone Marrow/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Membrane/metabolism , Epigenesis, Genetic , Genotype , HEK293 Cells , HL-60 Cells , Histone Demethylases/antagonists & inhibitors , Histones/chemistry , Humans , Leukocytes, Mononuclear/cytology
10.
Nat Commun ; 8(1): 1303, 2017 11 03.
Article in English | MEDLINE | ID: mdl-29101361

ABSTRACT

Genome-wide mapping of transcription factor binding is generally performed by chemical protein-DNA crosslinking, followed by chromatin immunoprecipitation and deep sequencing (ChIP-seq). Here we present the ChIP-seq technique based on photochemical crosslinking of protein-DNA interactions by high-intensity ultraviolet (UV) laser irradiation in living mammalian cells (UV-ChIP-seq). UV laser irradiation induces an efficient and instant formation of covalent "zero-length" crosslinks exclusively between nucleic acids and proteins that are in immediate contact, thus resulting in a "snapshot" of direct protein-DNA interactions in their natural environment. Here we show that UV-ChIP-seq, applied for genome-wide profiling of the sequence-specific transcriptional repressor B-cell lymphoma 6 (BCL6) in human diffuse large B-cell lymphoma (DLBCL) cells, produces sensitive and precise protein-DNA binding profiles, highly enriched with canonical BCL6 DNA sequence motifs. Using this technique, we also found numerous previously undetectable direct BCL6 binding sites, particularly in condensed, inaccessible areas of chromatin.


Subject(s)
Chromatin Immunoprecipitation/methods , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , Protein Binding , Binding Sites/genetics , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Photochemical Processes , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , Sequence Analysis, DNA , Ultraviolet Rays
11.
Nat Med ; 23(1): 69-78, 2017 01.
Article in English | MEDLINE | ID: mdl-27941792

ABSTRACT

In acute myeloid leukemia (AML), therapy resistance frequently occurs, leading to high mortality among patients. However, the mechanisms that render leukemic cells drug resistant remain largely undefined. Here, we identified loss of the histone methyltransferase EZH2 and subsequent reduction of histone H3K27 trimethylation as a novel pathway of acquired resistance to tyrosine kinase inhibitors (TKIs) and cytotoxic drugs in AML. Low EZH2 protein levels correlated with poor prognosis in AML patients. Suppression of EZH2 protein expression induced chemoresistance of AML cell lines and primary cells in vitro and in vivo. Low EZH2 levels resulted in derepression of HOX genes, and knockdown of HOXB7 and HOXA9 in the resistant cells was sufficient to improve sensitivity to TKIs and cytotoxic drugs. The endogenous loss of EZH2 expression in resistant cells and primary blasts from a subset of relapsed AML patients resulted from enhanced CDK1-dependent phosphorylation of EZH2 at Thr487. This interaction was stabilized by heat shock protein 90 (HSP90) and followed by proteasomal degradation of EZH2 in drug-resistant cells. Accordingly, inhibitors of HSP90, CDK1 and the proteasome prevented EZH2 degradation, decreased HOX gene expression and restored drug sensitivity. Finally, patients with reduced EZH2 levels at progression to standard therapy responded to the combination of bortezomib and cytarabine, concomitant with the re-establishment of EZH2 expression and blast clearance. These data suggest restoration of EZH2 protein as a viable approach to overcome treatment resistance in this AML patient population.


Subject(s)
Drug Resistance, Neoplasm/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Histones/metabolism , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Blotting, Western , Bortezomib/pharmacology , CDC2 Protein Kinase , Cell Line, Tumor , Cyclin-Dependent Kinases/metabolism , Cytarabine/pharmacology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HSP90 Heat-Shock Proteins/metabolism , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , Immunoprecipitation , Indoles/pharmacology , Leukemia, Myeloid, Acute/genetics , Male , Mass Spectrometry , Mice , Middle Aged , Neoplasm Transplantation , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Proteomics , Pyridones/pharmacology , Young Adult
12.
Nat Med ; 18(4): 605-11, 2012 Mar 11.
Article in English | MEDLINE | ID: mdl-22406747

ABSTRACT

Acute promyelocytic leukemia (APL), a cytogenetically distinct subtype of acute myeloid leukemia (AML), characterized by the t(15;17)-associated PML-RARA fusion, has been successfully treated with therapy utilizing all-trans-retinoic acid (ATRA) to differentiate leukemic blasts. However, among patients with non-APL AML, ATRA-based treatment has not been effective. Here we show that, through epigenetic reprogramming, inhibitors of lysine-specific demethylase 1 (LSD1, also called KDM1A), including tranylcypromine (TCP), unlocked the ATRA-driven therapeutic response in non-APL AML. LSD1 inhibition did not lead to a large-scale increase in histone 3 Lys4 dimethylation (H3K4(me2)) across the genome, but it did increase H3K4(me2) and expression of myeloid-differentiation-associated genes. Notably, treatment with ATRA plus TCP markedly diminished the engraftment of primary human AML cells in vivo in nonobese diabetic (NOD)-severe combined immunodeficient (SCID) mice, suggesting that ATRA in combination with TCP may target leukemia-initiating cells. Furthermore, initiation of ATRA plus TCP treatment 15 d after engraftment of human AML cells in NOD-SCID γ (with interleukin-2 (IL-2) receptor γ chain deficiency) mice also revealed the ATRA plus TCP drug combination to have a potent anti-leukemic effect that was superior to treatment with either drug alone. These data identify LSD1 as a therapeutic target and strongly suggest that it may contribute to AML pathogenesis by inhibiting the normal pro-differentiative function of ATRA, paving the way for new combinatorial therapies for AML.


Subject(s)
Cell Differentiation/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Tretinoin/therapeutic use , Animals , Antigens, CD34/metabolism , Apoptosis/drug effects , Apoptosis/physiology , CD11b Antigen/metabolism , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Drug Interactions , Enzyme Inhibitors/therapeutic use , Female , Flow Cytometry , Gene Expression Regulation/drug effects , Histone Demethylases/metabolism , Humans , Lysine/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Small Interfering/metabolism , Stem Cell Factor/metabolism , Time Factors , Transplants , Tranylcypromine/therapeutic use , Tretinoin/pharmacology
13.
Br J Haematol ; 154(4): 457-65, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21689085

ABSTRACT

The efficacy of tyrosine kinase (TK) inhibitors on non-cycling acute myeloid leukaemia (AML) cells, previously shown to have potent tumourigenic potential, is unknown. This pilot study describes the first attempt to characterize non-cycling cells from a small series of human FMS-like tyrosine kinase 3 (FLT3) mutation positive samples. CD34+ AML cells from patients with FLT3 mutation positive AML were cultured on murine stroma. In expansion cultures, non-cycling cells were found to retain CD34+ expression in contrast to dividing cells. Leukaemic gene rearrangements could be detected in non-cycling cells, indicating their leukaemic origin. Significantly, the FLT3-internal tandem duplication (ITD) mutation was found in the non-cycling fraction of four out of five cases. Exposure to the FLT3-directed inhibitor TKI258 clearly inhibited the growth of AML CD34+ cells in short-term cultures and colony-forming unit assays. Crucially, non-cycling cells were not eradicated, with the exception of one case, which exhibited exquisite sensitivity to the compound. Moreover, in longer-term cultures, TKI258-treated non-cycling cells showed no growth impairment compared to treatment-naive non-cycling cells. These findings suggest that non-cycling cells in AML may constitute a disease reservoir that is resistant to TK inhibition. Further studies with a larger sample size and other inhibitors are warranted.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Mutation , Quinolones/pharmacology , fms-Like Tyrosine Kinase 3/genetics , Adult , Animals , Antigens, CD34/metabolism , Cell Cycle/genetics , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Middle Aged , Pilot Projects , Protein-Tyrosine Kinases/antagonists & inhibitors , Tumor Cells, Cultured , Tumor Stem Cell Assay , Young Adult
14.
Proc Natl Acad Sci U S A ; 107(26): 11811-6, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20547842

ABSTRACT

Sin3A/B is a master transcriptional scaffold and corepressor that plays an essential role in the regulation of gene transcription and maintenance of chromatin structure, and its inappropriate recruitment has been associated with aberrant gene silencing in cancer. Sin3A/B are highly related, large, multidomian proteins that interact with a wide variety of transcription factors and corepressor components, and we examined whether disruption of the function of a specific domain could lead to epigenetic reprogramming and derepression of specific subsets of genes. To this end, we selected the Sin3A/B-paired amphipathic alpha-helices (PAH2) domain based on its established role in mediating the effects of a relatively small number of transcription factors containing a PAH2-binding motif known as the Sin3 interaction domain (SID). Here, we show that in both human and mouse breast cancer cells, the targeted disruption of Sin3 function by introduction of a SID decoy that interferes with PAH2 binding to SID-containing partner proteins reverted the silencing of genes involved in cell growth and differentiation. In particular, the SID decoy led to epigenetic reprogramming and reexpression of the important breast cancer-associated silenced genes encoding E-cadherin, estrogen receptor alpha, and retinoic acid receptor beta and impaired tumor growth in vivo. Interestingly, the SID decoy was effective in the triple-negative M.D. Anderson-Metastatic Breast-231 (MDA-MB-231) breast cancer cell line, restoring sensitivity to 17beta-estradiol, tamoxifen, and retinoids. Therefore, the development of small molecules that can block interactions between PAH2 and SID-containing proteins offers a targeted epigenetic approach for treating this type of breast cancer that may also have wider therapeutic implications.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Cell Differentiation/genetics , Cell Line, Tumor , Chromatin Assembly and Disassembly , DNA Primers/genetics , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Estrogen Receptor alpha/genetics , Female , Gene Targeting , Genetic Therapy , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Protein Binding , Protein Structure, Tertiary , Receptors, Retinoic Acid/genetics , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/chemistry , Sin3 Histone Deacetylase and Corepressor Complex
15.
Genes Chromosomes Cancer ; 46(9): 796-804, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17534929

ABSTRACT

The preferentially expressed antigen of melanoma (PRAME) is expressed at high levels in large fractions of human malignancies, e.g., acute myeloid leukemia. Therefore, PRAME is an important marker for diagnosis of various malignant diseases and a relevant parameter for monitoring minimal residual disease. It is supposed to be involved in tumorigenic processes. Because of these important aspects we investigated its transcriptional regulation in detail. Most relevant was a detailed DNA methylation analysis of the PRAME 5' region by genomic sequencing in correlation with PRAME expression in various human patient samples and cell lines. In combination with DNA-truncation/transfection experiments with respect to DNA methylation, we show that changes in the methylation pattern in defined parts of the regulatory regions of PRAME are sufficient for its upregulation in cells usually not expressing the gene.


Subject(s)
5' Flanking Region , Antigens, Neoplasm/metabolism , DNA Methylation , Leukemia, Myeloid, Acute/metabolism , Antigens, Neoplasm/genetics , Bone Marrow/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Myeloid, Acute/genetics , Promoter Regions, Genetic , Transcription, Genetic
16.
Eur J Cancer ; 42(16): 2807-14, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16978861

ABSTRACT

The gene PRAME (preferentially expressed antigen of melanoma) encodes an antigen recognised by autologous cytolytic T lymphocytes. The mRNA level of PRAME is used as a tumour marker due to its overexpression in various malignancies. Furthermore, it is known that the overexpression of genes encoding antiapoptotic proteins leads to the survival of leukaemic cells via exclusion of apoptosis. On the other hand, overexpression of genes encoding ABC transporters may lead to multi drug resistance (MDR). Therefore, we investigated whether there is a relationship between PRAME overexpression and the expression of apoptosis- and MDR-related genes in childhood de novo acute myelogenous leukaemia (AML) patient samples and, furthermore, whether this is a general or an AML-subtype specific event. Microarray analysis and real time quantitative PCR revealed that clinical samples showing PRAME upregulation are associated with a decreasing expression of genes coding for apoptotic proteins and an overexpression of genes encoding ABC transporters. Our results indicate that patients showing PRAME upregulation may have an increased risk of MDR induction.


Subject(s)
Antigens, Neoplasm/genetics , Genes, MDR/genetics , Leukemia, Myeloid/genetics , Acute Disease , Adolescent , Antigens, Neoplasm/metabolism , Apoptosis , Child , Child, Preschool , Drug Resistance, Multiple/genetics , Female , Gene Expression , Humans , Infant , Infant, Newborn , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/pathology , Male , Microarray Analysis , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...