Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Commun Signal ; 21(1): 354, 2023 12 15.
Article in English | MEDLINE | ID: mdl-38102712

ABSTRACT

BACKGROUND: Ephrin (EPH) receptors have been implicated in tumorigenesis and metastasis, but the functional understanding of mutations observed in human cancers is limited. We previously demonstrated reduced cell compartmentalisation for somatic EPHB1 mutations found in metastatic colorectal cancer cases. We therefore integrated pan-cancer and pan-EPH mutational data to prioritise recurrent EPHB1 mutations for functional studies to understand their contribution to cancer development and metastasis. METHODS: Here, 79,151 somatic mutations in 9,898 samples of 33 different tumour types were analysed with a bioinformatic pipeline to find 3D-mutated cluster pairs and hotspot mutations in EPH receptors. From these, 15 recurring EPHB1 mutations were stably expressed in colorectal cancer followed by confocal microscopy based in vitro compartmentalisation assays and phospho-proteome analysis. RESULTS: The 3D-protein structure-based bioinformatics analysis resulted in 63% EPHB1 mutants with compartmentalisation phenotypes vs 43% for hotspot mutations. Whereas the ligand-binding domain mutations C61Y, R90C, and R170W, the fibronectin domain mutation R351L, and the kinase domain mutation D762N displayed reduced to strongly compromised cell compartmentalisation, the kinase domain mutations R743W and G821R enhanced this phenotype. While mutants with reduced compartmentalisation also had reduced ligand induced receptor phosphorylation, the enhanced compartmentalisation was not linked to receptor phosphorylation level. Phosphoproteome mapping pinpointed the PI3K pathway and PIK3C2B phosphorylation in cells harbouring mutants with reduced compartmentalisation. CONCLUSIONS: This is the first integrative study of pan-cancer EPH receptor mutations followed by in vitro validation, a robust way to identify cancer-causing mutations, uncovering EPHB1 mutation phenotypes and demonstrating the utility of protein structure-based mutation analysis in characterization of novel cancer genes. Video Abstract.


Subject(s)
Colorectal Neoplasms , Phosphatidylinositol 3-Kinases , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Ligands , Mutation , Neoplasm Recurrence, Local , Receptor Protein-Tyrosine Kinases/metabolism
2.
Biochem Pharmacol ; 203: 115184, 2022 09.
Article in English | MEDLINE | ID: mdl-35872325

ABSTRACT

Loss of heterozygosity (LOH) is a hallmark feature of cancer genomes that reduces allelic variation, thereby creating tumor specific vulnerabilities which could be exploited for therapeutic purposes. We previously reported that loss of drug metabolic arylamine N-acetyltransferase 2 (NAT2) activity following LOH at 8p22 could be targeted for collateral lethality anticancer therapy in colorectal cancer (CRC). Here, we report a novel compound CBK034026C that exhibits specific toxicity towards CRC cells with high NAT2 activity. Connectivity Map analysis revealed that CBK034026C elicited a response pattern related to ATPase inhibitors. Similar to ouabain, a potent inhibitor of the Na+/K+-ATPase, CBK034026C activated the Nf-kB pathway. Further metabolomic profiling revealed downregulation of pathways associated with antioxidant defense and mitochondrial metabolism in CRC cells with high NAT2 activity, thereby weakening the protective response to oxidative stress induced by CBK034026C. The identification of a small molecule targeting metabolic vulnerabilities caused by NAT2 activity provides novel avenues for development of anticancer agents.


Subject(s)
Antineoplastic Agents , Arylamine N-Acetyltransferase , Colorectal Neoplasms , Acetyltransferases/genetics , Adenosine Triphosphatases , Alleles , Antineoplastic Agents/pharmacology , Arylamine N-Acetyltransferase/genetics , Arylamine N-Acetyltransferase/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Humans
3.
Mol Cell Oncol ; 7(4): 1759390, 2020.
Article in English | MEDLINE | ID: mdl-32944621

ABSTRACT

We show that N-acetyltransferase 2 (NAT2) loss of heterozygosity can be targeted in >4% of colorectal cancers with the use of a small molecule. We identify and describe the effect of a compound that impairs the growth of colorectal tumors with slow NAT2 activity by half when compared to wild-type.

4.
Nat Commun ; 11(1): 1308, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32161261

ABSTRACT

Cancer chemotherapy targeting frequent loss of heterozygosity events is an attractive concept, since tumor cells may lack enzymatic activities present in normal constitutional cells. To find exploitable targets, we map prevalent genetic polymorphisms to protein structures and identify 45 nsSNVs (non-synonymous small nucleotide variations) near the catalytic sites of 17 enzymes frequently lost in cancer. For proof of concept, we select the gastrointestinal drug metabolic enzyme NAT2 at 8p22, which is frequently lost in colorectal cancers and has a common variant with 10-fold reduced activity. Small molecule screening results in a cytotoxic kinase inhibitor that impairs growth of cells with slow NAT2 and decreases the growth of tumors with slow NAT2 by half as compared to those with wild-type NAT2. Most of the patient-derived CRC cells expressing slow NAT2 also show sensitivity to 6-(4-aminophenyl)-N-(3,4,5-trimethoxyphenyl)pyrazin-2-amine (APA) treatment. These findings indicate that the therapeutic index of anti-cancer drugs can be altered by bystander mutations affecting drug metabolic genes.


Subject(s)
Antineoplastic Agents/pharmacology , Arylamine N-Acetyltransferase/genetics , Colorectal Neoplasms/drug therapy , Loss of Heterozygosity , Protein Kinase Inhibitors/pharmacology , Alleles , Animals , Antineoplastic Agents/therapeutic use , Arylamine N-Acetyltransferase/metabolism , Bystander Effect/genetics , Case-Control Studies , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Female , HCT116 Cells , Humans , Isoenzymes/metabolism , Mice , Mice, Nude , Polymorphism, Genetic , Protein Kinase Inhibitors/therapeutic use , Small Molecule Libraries , Xenograft Model Antitumor Assays
5.
Sci Rep ; 10(1): 22436, 2020 12 31.
Article in English | MEDLINE | ID: mdl-33384440

ABSTRACT

Therapies targeting somatic bystander genetic events represent a new avenue for cancer treatment. We recently identified a subset of colorectal cancer (CRC) patients who are heterozygous for a wild-type and a low activity allele (NAT2*6) but lack the wild-type allele in their tumors due to loss of heterozygosity (LOH) at 8p22. These tumors were sensitive to treatment with a cytotoxic substrate of NAT2 (6-(4-aminophenyl)-N-(3,4,5-trimethoxyphenyl)pyrazin-2-amine, APA), and pointed to NAT2 loss being a therapeutically exploitable vulnerability of CRC tumors. To better estimate the total number of treatable CRC patients, we here determined whether tumor cells retaining also other NAT2 low activity variants after LOH respond to APA treatment. The prevalent low activity alleles NAT2*5 and NAT2*14, but not NAT2*7, were found to be low metabolizers with high sensitivity to APA. By analysis of two different CRC patient cohorts, we detected heterozygosity for NAT2 alleles targetable by APA, along with allelic imbalances pointing to LOH, in ~ 24% of tumors. Finally, to haplotype the NAT2 locus in tumor and patient-matched normal samples in a clinical setting, we develop and demonstrate a long-read sequencing based assay. In total, > 79.000 CRC patients per year fulfil genetic criteria for high sensitivity to a NAT2 LOH therapy and their eligibility can be assessed by clinical sequencing.


Subject(s)
Alleles , Antineoplastic Agents/therapeutic use , Arylamine N-Acetyltransferase/antagonists & inhibitors , Arylamine N-Acetyltransferase/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Enzyme Inhibitors/therapeutic use , Molecular Targeted Therapy , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Enzyme Inhibitors/pharmacology , Gene Frequency , Haplotypes , High-Throughput Nucleotide Sequencing , Humans , Phenotype , Single Molecule Imaging
6.
Oncotarget ; 8(58): 98646-98659, 2017 Nov 17.
Article in English | MEDLINE | ID: mdl-29228717

ABSTRACT

The chromatin modifier PRDM2/RIZ1 is inactivated by mutation in several forms of cancer and is a putative tumor suppressor gene. Frameshift mutations in the C-terminal region of PRDM2, affecting (A)8 or (A)9 repeats within exon 8, are found in one third of colorectal cancers with microsatellite instability, but the contribution of these mutations to colorectal tumorigenesis is unknown. To model somatic mutations in microsatellite unstable tumors, we devised a general approach to perform genome editing while stabilizing the mutated nucleotide repeat. We then engineered isogenic cell systems where the PRDM2 c.4467delA mutation in human HCT116 colorectal cancer cells was corrected to wild-type by genome editing. Restored PRDM2 increased global histone 3 lysine 9 dimethylation and reduced migration, anchorage-independent growth and tumor growth in vivo. Gene set enrichment analysis revealed regulation of several hallmark cancer pathways, particularly of epithelial-to-mesenchymal transition (EMT), with VIM being the most significantly regulated gene. These observations provide direct evidence that PRDM2 c.4467delA is a driver mutation in colorectal cancer and confirms PRDM2 as a cancer gene, pointing to regulation of EMT as a central aspect of its tumor suppressive action.

7.
Nucleic Acids Res ; 43(5): e30, 2015 Mar 11.
Article in English | MEDLINE | ID: mdl-25488813

ABSTRACT

The rapid discovery of potential driver mutations through large-scale mutational analyses of human cancers generates a need to characterize their cellular phenotypes. Among the techniques for genome editing, recombinant adeno-associated virus (rAAV)-mediated gene targeting is suited for knock-in of single nucleotide substitutions and to a lesser degree for gene knock-outs. However, the generation of gene targeting constructs and the targeting process is time-consuming and labor-intense. To facilitate rAAV-mediated gene targeting, we developed the first software and complementary automation-friendly vector tools to generate optimized targeting constructs for editing human protein encoding genes. By computational approaches, rAAV constructs for editing ~71% of bases in protein-coding exons were designed. Similarly, ~81% of genes were predicted to be targetable by rAAV-mediated knock-out. A Gateway-based cloning system for facile generation of rAAV constructs suitable for robotic automation was developed and used in successful generation of targeting constructs. Together, these tools enable automated rAAV targeting construct design, generation as well as enrichment and expansion of targeted cells with desired integrations.


Subject(s)
Dependovirus/genetics , Gene Targeting/methods , Genetic Vectors/genetics , Genome, Human/genetics , Computer Simulation , Databases, Genetic , Gene Expression Regulation , Gene Knock-In Techniques , Gene Knockout Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HCT116 Cells , Humans , Recombination, Genetic/genetics , Transfection/methods
8.
Nucleic Acids Res ; 40(17): 8440-8, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22753029

ABSTRACT

Ultraviolet (UV)-induced DNA damage causes an efficient block of elongating replication forks. The checkpoint kinase, CHK1 has been shown to stabilize replication forks following hydroxyurea treatment. Therefore, we wanted to test if the increased UV sensitivity caused by the unspecific kinase inhibitor caffeine--inhibiting ATM and ATR amongst other kinases--is explained by inability to activate the CHK1 kinase to stabilize replicative structures. For this, we used cells deficient in polymerase η (Polη), a translesion synthesis polymerase capable of properly bypassing the UV-induced cis-syn TT pyrimidine dimer, which blocks replication. These cells accumulate gaps behind progressing replication forks after UV exposure. We demonstrate that both caffeine and CHK1 inhibition, equally retards continuous replication fork elongation after UV treatment. Interestingly, we found more pronounced UV-sensitization by caffeine than with the CHK1 inhibitor in clonogenic survival experiments. Furthermore, we demonstrate an increased collapse of replicative structures after caffeine treatment, but not after CHK1 inhibition, in UV-irradiated cells. This demonstrates that CHK1 activity is not required for stabilization of gaps induced during replication of UV-damaged DNA. These data suggest that elongation and stabilization of replicative structures at UV-induced DNA damage are distinct mechanisms, and that CHK1 is only involved in replication elongation.


Subject(s)
DNA Damage , DNA Replication , Protein Kinases/metabolism , Ultraviolet Rays , Caffeine/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Transformed , Cell Survival , Checkpoint Kinase 1 , DNA Breaks, Double-Stranded , DNA Replication/drug effects , DNA Replication/radiation effects , DNA-Directed DNA Polymerase/deficiency , Humans , Protein Kinase Inhibitors/pharmacology , Signal Transduction/radiation effects
9.
BMC Res Notes ; 5: 87, 2012 Feb 06.
Article in English | MEDLINE | ID: mdl-22309575

ABSTRACT

BACKGROUND: The proliferating cell nuclear antigen (PCNA) is a key protein in the eukaryotic DNA replication and cell proliferation. Following the cloning and characterisation of the human PCNA gene, the question of the existence of pseudogenes in the human genome was raised. FINDINGS: In this short communication we summarise the existing information about the PCNA pseudogenes and critically assess their status. CONCLUSIONS: We propose the existence of at least four valid PCNA pseudogenes, PCNAP1, PCNAP2, LOC392454 and LOC390102. We would like to recommend assignment of a name for LOC392454 as "proliferating cell nuclear antigen pseudogene 3" (alias PCNAP3) and a name for LOC390102 as "proliferating cell nuclear antigen pseudogene 4" (alias PCNAP4). We prompt for more critical evaluation of the existence of a PCNA pseudogene, designated as PCNAP.

10.
PLoS One ; 6(5): e19492, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21573160

ABSTRACT

Homologous recombination (HR) is intricately associated with replication, transcription and DNA repair in all organisms studied. However, the interplay between all these processes occurring simultaneously on the same DNA molecule is still poorly understood. Here, we study the interplay between transcription and HR during ultraviolet light (UV)-induced DNA damage in mammalian cells. Our results show that inhibition of transcription with 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB) increases the number of UV-induced DNA lesions (γH2AX, 53BP1 foci formation), which correlates with a decrease in the survival of wild type or nucleotide excision repair defective cells. Furthermore, we observe an increase in RAD51 foci formation, suggesting HR is triggered in response to an increase in UV-induced DSBs, while inhibiting transcription. Unexpectedly, we observe that DRB fails to sensitise HR defective cells to UV treatment. Thus, increased RAD51 foci formation correlates with increased cell death, suggesting the existence of a futile HR repair of UV-induced DSBs which is linked to transcription inhibition.


Subject(s)
DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair/drug effects , Dichlororibofuranosylbenzimidazole/pharmacology , Recombination, Genetic/drug effects , Transcription, Genetic/drug effects , Ultraviolet Rays/adverse effects , Animals , Cell Line , Cricetinae , Cricetulus , DNA Repair/genetics , Flow Cytometry , Fluorescent Antibody Technique , Recombination, Genetic/genetics , Transcription, Genetic/genetics
11.
Mutat Res ; 706(1-2): 1-6, 2011 Jan 10.
Article in English | MEDLINE | ID: mdl-21074544

ABSTRACT

Transcription, replication and homologous recombination are intrinsically connected and it is well established that an increase of transcription is associated with an increase in homologous recombination. Here, we have studied how homologous recombination is affected during transcription inhibition by 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB), a compound that prevents activating phosphorylations of the RNA Pol II C-terminal domain. We identify that DRB triggers an increase in homologous recombination within the hprt gene as well as increasing RAD51 foci formation in mammalian cells. Furthermore, we find that DRB-induced transcriptional stress is associated with formation of the nuclear foci of the phosphorylated form of H2AX (γH2AX). We accounted that about 72% of RAD51 foci co-localized with the observed γH2AX foci. Interestingly, we find that XRCC3 mutated, homologous recombination defective cells are hypersensitive to the toxic effect of DRB and fail to form RAD51 foci. In conclusion, we show that DRB-induced transcription inhibition is associated with the formation of a lesion that triggers RAD51-dependent homologous recombination repair, required for survival under transcriptional stress.


Subject(s)
DNA Damage , DNA Repair/drug effects , Dichlororibofuranosylbenzimidazole/pharmacology , Recombination, Genetic/drug effects , Animals , CHO Cells , Cricetinae , Cricetulus , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Inhibitors/pharmacology , Histones/metabolism , Hypoxanthine Phosphoribosyltransferase/genetics , Microscopy, Confocal , Mutation , Phosphorylation/drug effects , Rad51 Recombinase/metabolism , Transcription, Genetic/drug effects
12.
Cancer Res ; 70(15): 6268-76, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20631063

ABSTRACT

Familial breast and ovarian cancers are often defective in homologous recombination (HR) due to mutations in the BRCA1 or BRCA2 genes. Cisplatin chemotherapy or poly(ADP-ribose) polymerase (PARP) inhibitors were tested for these tumors in clinical trials. In a screen for novel drugs that selectively kill BRCA2-defective cells, we identified 6-thioguanine (6TG), which induces DNA double-strand breaks (DSB) that are repaired by HR. Furthermore, we show that 6TG is as efficient as a PARP inhibitor in selectively killing BRCA2-defective tumors in a xenograft model. Spontaneous BRCA1-defective mammary tumors gain resistance to PARP inhibitors through increased P-glycoprotein expression. Here, we show that 6TG efficiently kills such BRCA1-defective PARP inhibitor-resistant tumors. We also show that 6TG could kill cells and tumors that have gained resistance to PARP inhibitors or cisplatin through genetic reversion of the BRCA2 gene. Although HR is reactivated in PARP inhibitor-resistant BRCA2-defective cells, it is not fully restored for the repair of 6TG-induced lesions. This is likely to be due to several recombinogenic lesions being formed after 6TG. We show that BRCA2 is also required for survival from mismatch repair-independent lesions formed by 6TG, which do not include DSBs. This suggests that HR is involved in the repair of 6TG-induced DSBs as well as mismatch repair-independent 6TG-induced DNA lesion. Altogether, our data show that 6TG efficiently kills BRCA2-defective tumors and suggest that 6TG may be effective in the treatment of advanced tumors that have developed resistance to PARP inhibitors or platinum-based chemotherapy.


Subject(s)
BRCA2 Protein/deficiency , Colonic Neoplasms/drug therapy , Enzyme Inhibitors/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Thioguanine/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis Regulatory Proteins , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Base Pair Mismatch , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , DNA Repair , Drug Synergism , Genes, BRCA2 , HCT116 Cells , Humans , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/genetics , Mice
13.
Biochem Soc Trans ; 37(Pt 3): 605-13, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19442257

ABSTRACT

Cancer is caused by genetic changes that often arise following failure to accurately replicate the DNA. PCNA (proliferating-cell nuclear antigen) forms a ring around the DNA to facilitate and control DNA replication. Emerging evidence suggests that PCNA is at the very heart of many essential cellular processes, such as DNA replication, repair of DNA damage, chromatin structure maintenance, chromosome segregation and cell-cycle progression. Progression of the DNA replication forks can be blocked by DNA lesions, formed either by endogenous damage or by exogenous agents, for instance anticancer drugs. Cellular response often results in change of PCNA function triggered either by specific post-translational modification of PCNA (i.e. ubiquitylation) or by exchange of its interaction partners. This puts PCNA in a central position in determining the fate of the replication fork. In the present article, we review PCNA modifications and interaction partners, and how those influence the course of events at replication forks, which ultimately determines both tumour progression as well as the outcome of anticancer treatment.


Subject(s)
Neoplasms/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Chromatin/metabolism , DNA Damage , DNA Repair/physiology , DNA Replication/physiology , Humans , Models, Molecular , Molecular Sequence Data , Neoplasms/pathology , Proliferating Cell Nuclear Antigen/chemistry , Proliferating Cell Nuclear Antigen/genetics , Protein Binding , Protein Conformation , Protein Processing, Post-Translational , Sequence Homology, Amino Acid
14.
J Cell Biochem ; 96(1): 126-36, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16052506

ABSTRACT

Host cell reactivation assay using Trioxsalen-crosslinked plasmid pEGFP-N1 showed that human cells were able to repair Trioxsalen interstrand crosslinks (ICL). To study the mechanism of this repair pathway, cells were transfected with the plasmids pEGFP-1, which did not contain the promoter of the egfp gene, and with pEGFP-G-, which did not contain the egfp gene. Neither of these plasmids alone was able to express the green fluorescent protein. After cotransfection with the two plasmids, 1%-2% of the cells developed fluorescent signal, which showed that recombination events had taken place in these cells to create DNA constructs containing the promoter and the gene properly aligned. When one or both of the plasmids were crosslinked with Trioxsalen, the recombination rate increased several fold. To identify the nuclear compartment where recombination takes place, cells were transfected with crosslinked pEGFP-N1 and the amount of plasmid DNA in the different nuclear fractions was determined. The results showed that Trioxsalen crosslinking increased the percentage of matrix attached plasmid DNA in a dose-dependent way. Immunoblotting experiments showed that after transfection with Trioxsalen crosslinked plasmids the homologous recombination protein Rad51 also associated with the nuclear matrix fraction. These studies provide a model system for investigating the precise molecular mechanisms that appear to couple repair of DNA ICL with nuclear matrix attachment.


Subject(s)
DNA Repair/physiology , DNA/metabolism , Nuclear Matrix/physiology , Animals , Cell Line , Cell Line, Tumor , Cricetinae , DNA/drug effects , Genes, Reporter , Humans , Nuclear Matrix/enzymology , Plasmids , Transfection , Trioxsalen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...