Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Nucleic Acids Res ; 52(3): 1136-1155, 2024 Feb 09.
Article in English | MEDLINE | ID: mdl-38038252

ABSTRACT

Maintaining chromatin integrity at the repetitive non-coding DNA sequences underlying centromeres is crucial to prevent replicative stress, DNA breaks and genomic instability. The concerted action of transcriptional repressors, chromatin remodelling complexes and epigenetic factors controls transcription and chromatin structure in these regions. The histone chaperone complex ATRX/DAXX is involved in the establishment and maintenance of centromeric chromatin through the deposition of the histone variant H3.3. ATRX and DAXX have also evolved mutually-independent functions in transcription and chromatin dynamics. Here, using paediatric glioma and pancreatic neuroendocrine tumor cell lines, we identify a novel ATRX-independent function for DAXX in promoting genome stability by preventing transcription-associated R-loop accumulation and DNA double-strand break formation at centromeres. This function of DAXX required its interaction with histone H3.3 but was independent of H3.3 deposition and did not reflect a role in the repression of centromeric transcription. DAXX depletion mobilized BRCA1 at centromeres, in line with BRCA1 role in counteracting centromeric R-loop accumulation. Our results provide novel insights into the mechanisms protecting the human genome from chromosomal instability, as well as potential perspectives in the treatment of cancers with DAXX alterations.


Subject(s)
Centromere , DNA Breaks, Double-Stranded , Molecular Chaperones , Nuclear Proteins , R-Loop Structures , X-linked Nuclear Protein , Child , Humans , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Centromere/metabolism , Chromatin , Co-Repressor Proteins/metabolism , DNA , Histones/genetics , Histones/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism
2.
Int J Mol Sci ; 24(12)2023 Jun 06.
Article in English | MEDLINE | ID: mdl-37372972

ABSTRACT

By generating protein diversity, alternative splicing provides an important oncogenic pathway. Isocitrate dehydrogenase (IDH) 1 and 2 mutations and 1p/19q co-deletion have become crucial for the novel molecular classification of diffuse gliomas, which also incorporates DNA methylation profiling. In this study, we have carried out a bioinformatics analysis to examine the impact of the IDH mutation, as well as the 1p/19q co-deletion and the glioma CpG island methylator phenotype (G-CIMP) status on alternative splicing in a cohort of 662 diffuse gliomas from The Cancer Genome Atlas (TCGA). We identify the biological processes and molecular functions affected by alternative splicing in the various glioma subgroups and provide evidence supporting the important contribution of alternative splicing in modulating epigenetic regulation in diffuse gliomas. Targeting the genes and pathways affected by alternative splicing might provide novel therapeutic opportunities against gliomas.


Subject(s)
Brain Neoplasms , Glioma , Humans , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Epigenesis, Genetic , Alternative Splicing , Glioma/genetics , Glioma/therapy , Mutation , Chromosome Aberrations , Phenotype , Isocitrate Dehydrogenase/genetics
3.
Cancers (Basel) ; 13(22)2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34830833

ABSTRACT

Despite their low incidence, pediatric high-grade gliomas (pHGGs), including diffuse intrinsic pontine gliomas (DIPGs), are the leading cause of mortality in pediatric neuro-oncology. Recurrent, mutually exclusive mutations affecting K27 (K27M) and G34 (G34R/V) in the N-terminal tail of histones H3.3 and H3.1 act as key biological drivers of pHGGs. Notably, mutations in H3.3 are frequently associated with mutations affecting ATRX and DAXX, which encode a chaperone complex that deposits H3.3 into heterochromatic regions, including telomeres. The K27M and G34R/V mutations lead to distinct epigenetic reprogramming, telomere maintenance mechanisms, and oncogenesis scenarios, resulting in distinct subgroups of patients characterized by differences in tumor localization, clinical outcome, as well as concurrent epigenetic and genetic alterations. Contrasting with our understanding of the molecular biology of pHGGs, there has been little improvement in the treatment of pHGGs, with the current mainstays of therapy-genotoxic chemotherapy and ionizing radiation (IR)-facing the development of tumor resistance driven by complex DNA repair pathways. Chromatin and nucleosome dynamics constitute important modulators of the DNA damage response (DDR). Here, we summarize the major DNA repair pathways that contribute to resistance to current DNA damaging agent-based therapeutic strategies and describe the telomere maintenance mechanisms encountered in pHGGs. We then review the functions of H3.3 and its chaperones in chromatin dynamics and DNA repair, as well as examining the impact of their mutation/alteration on these processes. Finally, we discuss potential strategies targeting DNA repair and epigenetic mechanisms as well as telomere maintenance mechanisms, to improve the treatment of pHGGs.

4.
Nucleic Acids Res ; 49(17): 9906-9925, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34500463

ABSTRACT

Replication-associated single-ended DNA double-strand breaks (seDSBs) are repaired predominantly through RAD51-mediated homologous recombination (HR). Removal of the non-homologous end-joining (NHEJ) factor Ku from resected seDSB ends is crucial for HR. The coordinated actions of MRE11-CtIP nuclease activities orchestrated by ATM define one pathway for Ku eviction. Here, we identify the pre-mRNA splicing protein XAB2 as a factor required for resistance to seDSBs induced by the chemotherapeutic alkylator temozolomide. Moreover, we show that XAB2 prevents Ku retention and abortive HR at seDSBs induced by temozolomide and camptothecin, via a pathway that operates in parallel to the ATM-CtIP-MRE11 axis. Although XAB2 depletion preserved RAD51 focus formation, the resulting RAD51-ssDNA associations were unproductive, leading to increased NHEJ engagement in S/G2 and genetic instability. Overexpression of RAD51 or RAD52 rescued the XAB2 defects and XAB2 loss was synthetically lethal with RAD52 inhibition, providing potential perspectives in cancer therapy.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Breaks, Double-Stranded , DNA End-Joining Repair/genetics , Ku Autoantigen/metabolism , RNA Splicing Factors/metabolism , Alkylating Agents/adverse effects , Alkylating Agents/pharmacology , Camptothecin/adverse effects , Camptothecin/pharmacology , Cell Line, Tumor , Endodeoxyribonucleases/metabolism , Glioblastoma/drug therapy , Homologous Recombination/genetics , Humans , MRE11 Homologue Protein/metabolism , RNA Interference , RNA Splicing Factors/genetics , RNA, Small Interfering/genetics , Rad51 Recombinase/metabolism , Rad52 DNA Repair and Recombination Protein/metabolism , Temozolomide/adverse effects , Temozolomide/pharmacology
5.
Cancers (Basel) ; 13(9)2021 May 07.
Article in English | MEDLINE | ID: mdl-34067180

ABSTRACT

BACKGROUND: Pediatric high-grade gliomas (pHGGs) are the leading cause of mortality in pediatric neuro-oncology, displaying frequent resistance to standard therapies. Profiling DNA repair and cell cycle gene expression has recently been proposed as a strategy to classify adult glioblastomas. To improve our understanding of the DNA damage response pathways that operate in pHGGs and the vulnerabilities that these pathways might expose, we sought to identify and characterize a specific DNA repair and cell-cycle gene expression signature of pHGGs. METHODS: Transcriptomic analyses were performed to identify a DNA repair and cell-cycle gene expression signature able to discriminate pHGGs (n = 6) from low-grade gliomas (n = 10). This signature was compared to related signatures already established. We used the pHGG signature to explore already transcriptomic datasets of DIPGs and sus-tentorial pHGGs. Finally, we examined the expression of key proteins of the pHGG signature in 21 pHGG diagnostic samples and nine paired relapses. Functional inhibition of one DNA repair factor was carried out in four patients who derived H3.3 K27M mutant cell lines. RESULTS: We identified a 28-gene expression signature of DNA repair and cell cycle that clustered pHGGs cohorts, in particular sus-tentorial locations, in two groups. Differential protein expression levels of PARP1 and XRCC1 were associated to TP53 mutations and TOP2A amplification and linked significantly to the more radioresistant pHGGs displaying the worst outcome. Using patient-derived cell lines, we showed that the PARP-1/XRCC1 expression balance might be correlated with resistance to PARP1 inhibition. CONCLUSION: We provide evidence that PARP1 overexpression, associated to XRCC1 expression, TP53 mutations, and TOP2A amplification, is a new theranostic and potential therapeutic target.

6.
Cancers (Basel) ; 12(9)2020 Sep 10.
Article in English | MEDLINE | ID: mdl-32927769

ABSTRACT

Resistance to chemotherapy by temozolomide (TMZ) is a major cause of glioblastoma (GBM) recurrence. So far, attempts to characterize factors that contribute to TMZ sensitivity have largely focused on protein-coding genes, and failed to provide effective therapeutic targets. Long noncoding RNAs (lncRNAs) are essential regulators of epigenetic-driven cell diversification, yet, their contribution to the transcriptional response to drugs is less understood. Here, we performed RNA-seq and small RNA-seq to provide a comprehensive map of transcriptome regulation upon TMZ in patient-derived GBM stem-like cells displaying different drug sensitivity. In a search for regulatory mechanisms, we integrated thousands of molecular associations stored in public databases to generate a background "RNA interactome". Our systems-level analysis uncovered a coordinated program of TMZ response reflected by regulatory circuits that involve transcription factors, mRNAs, miRNAs, and lncRNAs. We discovered 22 lncRNAs involved in regulatory loops and/or with functional relevance in drug response and prognostic value in gliomas. Thus, the investigation of TMZ-induced gene networks highlights novel RNA-based predictors of chemosensitivity in GBM. The computational modeling used to identify regulatory circuits underlying drug response and prioritizing gene candidates for functional validation is applicable to other datasets.

7.
Cancer Res ; 79(6): 1226-1238, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30674534

ABSTRACT

Inevitable tumor recurrence and a poor median survival are frustrating reminders of the inefficacy of our current standard of care for patients with newly diagnosed glioblastoma (GBM), which includes surgery followed by radiotherapy and chemotherapy with the DNA alkylating agent temozolomide. Because resistance to genotoxic damage is achieved mainly through execution of the DNA damage response (DDR) and DNA repair pathways, knowledge of the changes in DNA repair and cell-cycle gene expression that occur during tumor development might help identify new targets and improve treatment. Here, we performed a gene expression analysis targeting components of the DNA repair and cell-cycle machineries in cohorts of paired tumor samples (i.e., biopsies from the same patient obtained at the time of primary tumor operation and at recurrence) from patients treated with radiotherapy or radiotherapy plus temozolomide. We identified and validated a 27-gene signature that resulted in the classification of GBM specimens into three groups, two of which displayed inverse expression profiles. Each group contained primary and recurrent samples, and the tumor at relapse frequently displayed a gene expression profile different from that of the matched primary biopsy. Within the groups that exhibited opposing gene expression profiles, the expression pattern of the gene signature at relapse was linked to progression-free survival. We provide experimental evidence that our signature exposes group-specific vulnerabilities against genotoxicants and inhibitors of the cell cycle and DDR, with the prospect of personalized therapeutic strategies.Significance: These findings suggest that classification of GBM tumors based on a DNA repair and cell-cycle gene expression signature exposes vulnerabilities to standard-of-care therapies and offers the potential for personalized therapeutic strategies.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Cell Cycle Proteins/genetics , DNA Repair Enzymes/genetics , Glioblastoma/genetics , Neoplasm Recurrence, Local/genetics , Adult , Aged , Apoptosis , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Proliferation , Cohort Studies , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/radiotherapy , Prognosis , Survival Rate , Tumor Cells, Cultured
8.
Trends Biotechnol ; 37(1): 38-55, 2019 01.
Article in English | MEDLINE | ID: mdl-30177380

ABSTRACT

High-throughput genetic screens interfering with gene expression are invaluable tools to identify gene function and phenotype-to-genotype interactions. Implementing such screens in the laboratory is challenging, and the choice between currently available technologies based on RNAi and CRISPR/Cas9 (CRISPR-associated protein 9) is not trivial. Identifying reliable candidate hits requires a streamlined experimental setup adjusted to the specific biological question. Here, we provide a critical assessment of the various RNAi/CRISPR approaches to pooled screens and discuss their advantages and pitfalls. We specify a set of best practices for key parameters enabling a reproducible screen and provide a detailed overview of analysis methods and repositories for identifying the best candidate gene hits.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Gene Knock-In Techniques/methods , Gene Knockdown Techniques/methods , Gene Knockout Techniques/methods , Genetic Association Studies , Genetic Testing/methods , RNA Interference , Computational Biology/methods , High-Throughput Screening Assays
9.
Nucleic Acids Res ; 47(3): 1051-1069, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30590707

ABSTRACT

The histone H3 variant CENP-A confers epigenetic identity to the centromere and plays crucial roles in the assembly and function of the kinetochore, thus ensuring proper segregation of our chromosomes. CENP-A containing nucleosomes exhibit unique structural specificities and lack the complex profile of gene expression-associated histone posttranslational modifications found in canonical histone H3 and the H3.3 variant. CENP-A mislocalization into noncentromeric regions resulting from its overexpression leads to chromosomal segregation aberrations and genome instability. Overexpression of CENP-A is a feature of many cancers and is associated with malignant progression and poor outcome. The recent years have seen impressive progress in our understanding of the mechanisms that orchestrate CENP-A deposition at native centromeres and ectopic loci. They have witnessed the description of novel, heterotypic CENP-A/H3.3 nucleosome particles and the exploration of the phenotypes associated with the deregulation of CENP-A and its chaperones in tumor cells. Here, we review the structural specificities of CENP-A nucleosomes, the epigenetic features that characterize the centrochromatin and the mechanisms and factors that orchestrate CENP-A deposition at centromeres. We then review our knowledge of CENP-A ectopic distribution, highlighting experimental strategies that have enabled key discoveries. Finally, we discuss the implications of deregulated CENP-A in cancer.


Subject(s)
Centromere Protein A/metabolism , Centromere/metabolism , Chromatin/metabolism , Neoplasms/genetics , Animals , Centromere/chemistry , Epigenesis, Genetic , Humans , Neoplasms/metabolism , Nucleosomes/chemistry , Nucleosomes/metabolism
10.
Mutat Res Rev Mutat Res ; 769: 19-35, 2016.
Article in English | MEDLINE | ID: mdl-27543314

ABSTRACT

Despite surgical resection and genotoxic treatment with ionizing radiation and the DNA alkylating agent temozolomide, glioblastoma remains one of the most lethal cancers, due in great part to the action of DNA repair mechanisms that drive resistance and tumor relapse. Understanding the molecular details of these mechanisms and identifying potential pharmacological targets have emerged as vital tasks to improve treatment. In this review, we introduce the various cellular systems and animal models that are used in studies of DNA repair in glioblastoma. We summarize recent progress in our knowledge of the pathways and factors involved in the removal of DNA lesions induced by ionizing radiation and temozolomide. We introduce the therapeutic strategies relying on DNA repair inhibitors that are currently being tested in vitro or in clinical trials, and present the challenges raised by drug delivery across the blood brain barrier as well as new opportunities in this field. Finally, we review the genetic and epigenetic alterations that help shape the DNA repair makeup of glioblastoma cells, and discuss their potential therapeutic impact and implications for personalized therapy.


Subject(s)
Brain Neoplasms/genetics , DNA Repair , Glioblastoma/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers , Brain Neoplasms/diagnosis , Brain Neoplasms/metabolism , Brain Neoplasms/therapy , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Disease Models, Animal , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genetic Therapy , Glioblastoma/diagnosis , Glioblastoma/metabolism , Glioblastoma/therapy , Humans , Radiotherapy , Signal Transduction
11.
Cancer Med ; 3(2): 322-36, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24497500

ABSTRACT

Cigarette smoking is the major cause of cancers of the respiratory tract, including non-small cell lung cancer (NSCLC) and head and neck cancer (HNC). In order to better understand carcinogenesis of the lung and upper airways, we have compared the gene expression profiles of tumor-distant, histologically normal bronchial biopsy specimens obtained from current smokers with NSCLC or HNC (SC, considered as a single group), as well as nonsmokers (NS) and smokers without cancer (SNC). RNA from a total of 97 biopsies was used for gene expression profiling (Affymetrix HG-U133 Plus 2.0 array). Differentially expressed genes were used to compare NS, SNC, and SC, and functional analysis was carried out using Ingenuity Pathway Analysis (IPA). Smoking-related cancer of the respiratory tract was found to affect the expression of genes encoding xenobiotic biotransformation proteins, as well as proteins associated with crucial inflammation/immunity pathways and other processes that protect the airway from the chemicals in cigarette smoke or contribute to carcinogenesis. Finally, we used the prediction analysis for microarray (PAM) method to identify gene signatures of cigarette smoking and cancer, and uncovered a 15-gene signature that distinguished between SNC and SC with an accuracy of 83%. Thus, gene profiling of histologically normal bronchial biopsy specimens provided insight into cigarette-induced carcinogenesis of the respiratory tract and gene signatures of cancer in smokers.


Subject(s)
Bronchi/physiology , Carcinoma, Non-Small-Cell Lung/genetics , Head and Neck Neoplasms/genetics , Lung Neoplasms/genetics , Smoking/genetics , Adult , Aged , Bronchi/metabolism , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Smoking/adverse effects
12.
Carcinogenesis ; 32(3): 271-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21112961

ABSTRACT

The tumor suppressor gene TP53, encoding p53, is expressed as several transcripts. The fully spliced p53 (FSp53) transcript encodes the canonical p53 protein. The alternatively spliced p53I2 transcript retains intron 2 and encodes Δ40p53 (or ΔNp53), an isoform lacking first 39 N-terminal residues corresponding to the main transactivation domain. We demonstrate the formation of G-quadruplex structures (G4) in a GC-rich region of intron 3 that modulates the splicing of intron 2. First, we show the formation of G4 in synthetic RNAs encompassing intron 3 sequences by ultraviolet melting, thermal difference spectra and circular dichroism spectroscopy. These observations are confirmed by detection of G4-induced reverse transcriptase elongation stops in synthetic RNA of intron 3. In this region, p53 pre-messenger RNA (mRNA) contains a succession of short exons (exons 2 and 3) and introns (introns 2 and 4) covering a total of 333 bp. Site-directed mutagenesis of G-tracts putatively involved in G4 formation decreased by ~30% the excision of intron 2 in a green fluorescent protein-reporter splicing assay. Moreover, treatment of lymphoblastoid cells with 360A, a synthetic ligand that binds to single-strand G4 structures, increases the formation of FSp53 mRNA and decreases p53I2 mRNA expression. These results indicate that G4 structures in intron 3 regulate the splicing of intron 2, leading to differential expression of transcripts encoding distinct p53 isoforms.


Subject(s)
Alternative Splicing , G-Quadruplexes , RNA, Messenger/genetics , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/genetics , Base Sequence , Circular Dichroism , Exons/genetics , Green Fluorescent Proteins/metabolism , Humans , Introns/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Isoforms , RNA-Directed DNA Polymerase/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
13.
Mol Cancer ; 9: 115, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20487546

ABSTRACT

BACKGROUND: Chronic lymphocytic leukemia (CLL) cells are often affected by genomic aberrations targeting key regulatory genes. Although fludarabine is the standard first line therapy to treat CLL, only few data are available about the resistance of B cells to this purine nucleoside analog in vivo. Here we sought to increase our understanding of fludarabine action and describe the mechanisms leading to resistance in vivo. We performed an analysis of genomic aberrations, gene expression profiles, and microRNAs expression in CLL blood B lymphocytes isolated during the course of patients' treatment with fludarabine. RESULTS: In sensitive patients, the differentially expressed genes we identified were mainly involved in p53 signaling, DNA damage response, cell cycle and cell death. In resistant patients, uncommon genomic abnormalities were observed and the resistance toward fludarabine could be characterized based on the expression profiles of genes implicated in lymphocyte proliferation, DNA repair, and cell growth and survival. Of particular interest in some patients was the amplification of MYC (8q) observed both at the gene and transcript levels, together with alterations of myc-transcriptional targets, including genes and miRNAs involved in the regulation of cell cycle and proliferation. Differential expression of the sulfatase SULF2 and of miR-29a, -181a, and -221 was also observed between resistant and sensitive patients before treatment. These observations were further confirmed on a validation cohort of CLL patients treated with fludarabine in vitro. CONCLUSION: In the present study we identified genes and miRNAs that may predict clinical resistance of CLL to fludarabine, and describe an interesting oncogenic mechanism in CLL patients resistant to fludarabine by which the complete MYC-specific regulatory network was altered (DNA and RNA levels, and transcriptional targets). These results should prove useful for understanding and overcoming refractoriness to fludarabine and also for predicting the clinical outcome of CLL patients before or early during their treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , MicroRNAs/genetics , Vidarabine/analogs & derivatives , Aged , Comparative Genomic Hybridization , Female , Gene Expression/drug effects , Genes, myc/genetics , Humans , In Situ Hybridization, Fluorescence , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics , Vidarabine/therapeutic use
14.
Mol Cancer Ther ; 9(5): 1349-60, 2010 May.
Article in English | MEDLINE | ID: mdl-20406947

ABSTRACT

Clinical trials have shown activity of the isotype-selective histone deacetylase (HDAC) inhibitor MGCD0103 in different hematologic malignancies. There are data to support the use of HDAC inhibitors in association with other cancer therapies. To propose a rational combination therapy, it is necessary to depict the molecular basis behind the cytotoxic effect of MGCD0103. In this study, we found that MGCD0103 was substantially more toxic in neoplastic B cells relative to normal cells, and we described the death pathways activated by MGCD0103 in B-cell chronic lymphocytic leukemia (CLL) cells from 32 patients. MGCD0103 decreased the expression of Mcl-1 and induced translocation of Bax to the mitochondria, mitochondrial depolarization, and release of cytochrome c in the cytosol. Caspase processing in the presence of the caspase inhibitor Q-VD-OPh and time course experiments showed that caspase-9 was the apical caspase. Thus, MGCD0103 induced the intrinsic pathway of apoptosis in CLL cells. Moreover, MGCD0103 treatment resulted in the activation of a caspase cascade downstream of caspase-9, caspase-dependent amplification of mitochondrial depolarization, activation of calpain, and Bax cleavage. We propose a model whereby the intrinsic pathway of apoptosis triggered by MGCD0103 in CLL is associated with a mitochondrial death amplification loop.


Subject(s)
Apoptosis/drug effects , Benzamides/pharmacology , Caspases/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Pyrimidines/pharmacology , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Cells, Cultured , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Female , Histone Deacetylase Inhibitors/pharmacology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Middle Aged , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Signal Transduction/drug effects , Up-Regulation/drug effects
15.
Nucleic Acids Res ; 35(19): 6571-87, 2007.
Article in English | MEDLINE | ID: mdl-17905820

ABSTRACT

The RDM1 gene encodes a RNA recognition motif (RRM)-containing protein involved in the cellular response to the anti-cancer drug cisplatin in vertebrates. We previously reported a cDNA encoding the full-length human RDM1 protein. Here, we describe the identification of 11 human cDNAs encoding RDM1 protein isoforms. This repertoire is generated by alternative pre-mRNA splicing and differential usage of two translational start sites, resulting in proteins with long or short N-terminus and a great diversity in the exonic composition of their C-terminus. By using tagged proteins and fluorescent microscopy, we examined the subcellular distribution of full-length RDM1 (renamed RDM1alpha), and other RDM1 isoforms. We show that RDM1alpha undergoes subcellular redistribution and nucleolar accumulation in response to proteotoxic stress and mild heat shock. In unstressed cells, the long N-terminal isoforms displayed distinct subcellular distribution patterns, ranging from a predominantly cytoplasmic to almost exclusive nuclear localization, suggesting functional differences among the RDM1 proteins. However, all isoforms underwent stress-induced nucleolar accumulation. We identified nuclear and nucleolar localization determinants as well as domains conferring cytoplasmic retention to the RDM1 proteins. Finally, RDM1 null chicken DT40 cells displayed an increased sensitivity to heat shock, compared to wild-type (wt) cells, suggesting a function for RDM1 in the heat-shock response.


Subject(s)
Cell Nucleolus/chemistry , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Heat-Shock Response , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Alternative Splicing , Animals , Base Sequence , Cell Line , Cell Nucleus/chemistry , Chickens , Cysteine Proteinase Inhibitors/pharmacology , Cytoplasm/chemistry , DNA-Binding Proteins/chemistry , Dactinomycin/pharmacology , Exons , Gene Deletion , Humans , Molecular Sequence Data , Nucleic Acid Synthesis Inhibitors/pharmacology , Proteasome Inhibitors , Protein Isoforms/analysis , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Structure, Tertiary , RNA, Messenger/metabolism , Transcription, Genetic/drug effects
16.
Biochem Biophys Res Commun ; 344(1): 87-94, 2006 May 26.
Article in English | MEDLINE | ID: mdl-16630539

ABSTRACT

RDM1 (RAD52 Motif 1) is a vertebrate protein involved in the cellular response to the anti-cancer drug cisplatin. In addition to an RNA recognition motif, RDM1 contains a small amino acid motif, named RD motif, which it shares with the recombination and repair protein, RAD52. RDM1 binds to single- and double-stranded DNA, and recognizes DNA distortions induced by cisplatin adducts in vitro. Here, we have performed an in-depth analysis of the nucleic acid-binding properties of RDM1 using gel-shift assays and electron microscopy. We show that RDM1 possesses acidic pH-dependent DNA-binding activity and that it binds RNA as well as DNA, and we present evidence from competition gel-shift experiments that RDM1 may be capable of discrimination between the two nucleic acids. Based on reported studies of RAD52, we have generated an RDM1 variant mutated in its RD motif. We find that the L119GF --> AAA mutation affects the mode of RDM1 binding to single-stranded DNA.


Subject(s)
DNA, Single-Stranded/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA/chemistry , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Apoptosis , Cells, Cultured , Chickens , DNA, Single-Stranded/ultrastructure , DNA-Binding Proteins/ultrastructure , Electrophoretic Mobility Shift Assay , Hydrogen-Ion Concentration , Molecular Sequence Data , Mutation , RNA/ultrastructure , RNA-Binding Proteins/ultrastructure
17.
J Biol Chem ; 280(10): 9225-35, 2005 Mar 11.
Article in English | MEDLINE | ID: mdl-15611051

ABSTRACT

A variety of cellular proteins has the ability to recognize DNA lesions induced by the anti-cancer drug cisplatin, with diverse consequences on their repair and on the therapeutic effectiveness of this drug. We report a novel gene involved in the cell response to cisplatin in vertebrates. The RDM1 gene (for RAD52 Motif 1) was identified while searching databases for sequences showing similarities to RAD52, a protein involved in homologous recombination and DNA double-strand break repair. Ablation of RDM1 in the chicken B cell line DT40 led to a more than 3-fold increase in sensitivity to cisplatin. However, RDM1-/- cells were not hypersensitive to DNA damages caused by ionizing radiation, UV irradiation, or the alkylating agent methylmethane sulfonate. The RDM1 protein displays a nucleic acid binding domain of the RNA recognition motif (RRM) type. By using gel-shift assays and electron microscopy, we show that purified, recombinant chicken RDM1 protein interacts with single-stranded DNA as well as double-stranded DNA, on which it assembles filament-like structures. Notably, RDM1 recognizes DNA distortions induced by cisplatin-DNA adducts in vitro. Finally, human RDM1 transcripts are abundant in the testis, suggesting a possible role during spermatogenesis.


Subject(s)
Cisplatin/pharmacology , DNA-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Survival , Chickens , Cloning, Molecular , DNA Repair , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Expressed Sequence Tags , Humans , Kinetics , Markov Chains , Models, Molecular , Molecular Sequence Data , Protein Conformation , Recombination, Genetic , Sequence Alignment , Sequence Deletion , Sequence Homology, Amino Acid , Vertebrates
SELECTION OF CITATIONS
SEARCH DETAIL
...