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










Publication year range
1.
JCO Precis Oncol ; 8: e2300289, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38412387

ABSTRACT

PURPOSE: Cell-free circulating tumor DNA (ctDNA) has shown its potential as a quantitative biomarker for longitudinal monitoring of response to anticancer therapies. However, ctDNA dynamics have not been studied in patients with heavily pretreated, advanced solid tumors, for whom therapeutic responses can be weak. We investigated whether changes in ctDNA could predict clinical outcomes in such a cohort treated with combined poly(ADP-ribose) polymerase/vascular endothelial growth factor receptor inhibitor therapy. MATERIALS AND METHODS: Patients with metastatic pancreatic ductal adenocarcinoma (PDAC), triple-negative breast cancer (TNBC), small-cell lung cancer (SCLC), or non-small-cell lung cancer (NSCLC) received up to 7 days of cediranib 30 mg orally once daily monotherapy lead-in followed by addition of olaparib 200 mg orally twice daily. Patients had progressed on a median of three previous lines of therapy. Plasma samples were collected before and after cediranib monotherapy lead-in and on combination therapy at 7 days, 28 days, and every 28 days thereafter. ctDNA was quantified from plasma samples using a multigene mutation-based assay. Radiographic assessment was performed every 8 weeks. RESULTS: ctDNA measurements were evaluable in 63 patients. The median baseline ctDNA variant allele fractions (VAFs) were 20%, 28%, 27%, and 34% for PDAC, TNBC, SCLC, and NSCLC, respectively. No association was observed between baseline VAF and radiographic response, progression-free survival, or overall survival (OS). Similarly, no association was found between ctDNA decline and radiographic response or survival. However, an increase in ctDNA at 56 days of combination therapy was associated with disease progression and inferior OS in a landmark analysis. CONCLUSION: ctDNA levels or dynamics did not correlate with radiographic response or survival outcomes in patients with advanced metastatic malignancies treated with olaparib and cediranib.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Circulating Tumor DNA , Lung Neoplasms , Pancreatic Neoplasms , Triple Negative Breast Neoplasms , Humans , Circulating Tumor DNA/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Poly(ADP-ribose) Polymerases/therapeutic use , Vascular Endothelial Growth Factor A/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Biomarkers, Tumor/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics
2.
Proc Natl Acad Sci U S A ; 117(4): 2084-2091, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31932421

ABSTRACT

BRCA1 promotes error-free, homologous recombination-mediated repair (HRR) of DNA double-stranded breaks (DSBs). When excessive and uncontrolled, BRCA1 HRR activity promotes illegitimate recombination and genome disorder. We and others have observed that the BRCA1-associated protein RAP80 recruits BRCA1 to postdamage nuclear foci, and these chromatin structures then restrict the amplitude of BRCA1-driven HRR. What remains unclear is how this process is regulated. Here we report that both BRCA1 poly-ADP ribosylation (PARsylation) and the presence of BRCA1-bound RAP80 are critical for the normal interaction of BRCA1 with some of its partners (e.g., CtIP and BACH1) that are also known components of the aforementioned focal structures. Surprisingly, the simultaneous loss of RAP80 and failure therein of BRCA1 PARsylation results in the dysregulated accumulation in these foci of BRCA1 complexes. This in turn is associated with the intracellular development of a state of hyper-recombination and gross chromosomal disorder. Thus, physiological RAP80-BRCA1 complex formation and BRCA1 PARsylation contribute to the kinetics by which BRCA1 HRR-sustaining complexes normally concentrate in nuclear foci. These events likely contribute to aneuploidy suppression.


Subject(s)
BRCA1 Protein/metabolism , DNA-Binding Proteins/metabolism , Histone Chaperones/metabolism , Recombinational DNA Repair , BRCA1 Protein/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Chromosomes/genetics , Chromosomes/metabolism , DNA Damage , DNA-Binding Proteins/genetics , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism , Histone Chaperones/genetics , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Poly ADP Ribosylation , Protein Binding
3.
Cancer Discov ; 4(12): 1430-47, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25252691

ABSTRACT

UNLABELLED: BRCA1 promotes homologous recombination-mediated DNA repair (HRR). However, HRR must be tightly regulated to prevent illegitimate recombination. We previously found that BRCA1 HRR function is regulated by the RAP80 complex, but the mechanism was unclear. We have now observed that PARP1 interacts with and poly-ADP-ribosylates (aka PARsylates) BRCA1. PARsylation is directed at the BRCA1 DNA binding domain and downmodulates its function. Moreover, RAP80 contains a poly-ADP-ribose-interacting domain that binds PARsylated BRCA1 and helps to maintain the stability of PARP1-BRCA1-RAP80 complexes. BRCA1 PARsylation is a key step in BRCA1 HRR control. When BRCA1 PARsylation is defective, it gives rise to excessive HRR and manifestations of genome instability. BRCA1 PARsylation and/or RAP80 expression is defective in a subset of sporadic breast cancer cell lines and patient-derived tumor xenograft models. These observations are consistent with the possibility that such defects, when chronic, contribute to tumor development in BRCA1+/+ individuals. SIGNIFICANCE: We propose a model that describes how BRCA1 functions to both support and restrict HRR. BRCA1 PARsylation is a key event in this process, failure of which triggers hyper-recombination and chromosome instability. Thus, hyperfunctioning BRCA1 can elicit genomic abnormalities similar to those observed in the absence of certain BRCA1 functions.


Subject(s)
BRCA1 Protein/metabolism , DNA Repair , Homologous Recombination , Poly(ADP-ribose) Polymerases/metabolism , Amino Acid Sequence , BRCA1 Protein/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chromosomes, Human , DNA Damage , DNA-Binding Proteins , Female , Gene Expression , Genomic Instability , HeLa Cells , Histone Chaperones , Humans , Models, Biological , Multiprotein Complexes/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptide Fragments , Poly (ADP-Ribose) Polymerase-1 , Protein Binding , Protein Interaction Domains and Motifs , Protein Stability , Protein Subunits/metabolism , Signal Transduction
4.
Genes Dev ; 27(20): 2274-91, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24142877

ABSTRACT

Endogenous BRCA1 p220 expression peaks in S and G2 when it is activated, and the protein participates in certain key DNA damage responses. In contrast, its expression is markedly reduced in G0/G1. While variations in transcription represent a significant part of p220 expression control, there is at least one other relevant process. We found that a microRNA, miR-545, that is expressed throughout the cell cycle down-modulates endogenous p220 mRNA and protein abundance directly in both G0/G1 and S/G2. When miR-545 function was inhibited by a specific antagomir, endogenous p220 expression increased in G0/G1, and aberrant p220-associated DNA damage responses and de novo DNA strand breaks accumulated. Analogous results were observed upon inhibition of miR-545 function in S/G2. Both sets of antagomir effects were mimicked by infecting cells with a p220 cDNA-encoding adenoviral vector. Thus, strand breaks were a product of p220 overexpression, and their prevention by miR-545 depends on its modulation of p220 expression. Breaks were also dependent on aberrant, overexpressed p220-driven recruitment of RAD51 to either spontaneously arising or mutagen-based DNA damage sites. Hence, when its level is not physiologically maintained, endogenous p220 aberrantly directs at least one DNA repair protein, RAD51, to damage sites, where their action contributes to the development of de novo DNA damage. Thus, like its loss, a surfeit of endogenous p220 function represents a threat to genome integrity.


Subject(s)
BRCA1 Protein/genetics , Cell Cycle/physiology , DNA Damage/genetics , Gene Expression Regulation , BRCA1 Protein/metabolism , Binding Sites , Cell Line, Tumor , DNA Damage/radiation effects , DNA Repair , HEK293 Cells , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Binding , RNA, Messenger/metabolism , Rad51 Recombinase/metabolism , Ultraviolet Rays
5.
Mol Cell ; 48(5): 723-33, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23122415

ABSTRACT

Chromatin remodeling during DNA double-strand break (DSB) repair is required to facilitate access to and repair of DSBs. This remodeling requires increased acetylation of histones and a shift in nucleosome organization to create open, relaxed chromatin domains. However, the underlying mechanism driving changes in nucleosome structure at DSBs is poorly defined. Here, we demonstrate that histone H2A.Z is exchanged onto nucleosomes at DSBs by the p400 remodeling ATPase. H2A.Z exchange at DSBs shifts the chromatin to an open conformation and is required for acetylation and ubiquitination of histones and for loading of the brca1 complex. H2A.Z exchange also restricts single-stranded DNA production by nucleases and is required for loading of the Ku70/Ku80 DSB repair protein. H2A.Z exchange therefore promotes specific patterns of histone modification and reorganization of the chromatin architecture, leading to the assembly of a chromatin template that is an efficient substrate for the DSB repair machinery.


Subject(s)
Chromatin Assembly and Disassembly , DNA Breaks, Double-Stranded , DNA Repair , Histones/metabolism , Nucleosomes/metabolism , Acetylation , Adenosine Triphosphatases/metabolism , Antigens, Nuclear/metabolism , Binding Sites , Binding, Competitive , Carrier Proteins/metabolism , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Radiation , Endodeoxyribonucleases , HEK293 Cells , HeLa Cells , Histones/chemistry , Histones/genetics , Humans , Ku Autoantigen , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/radiation effects , RNA Interference , Time Factors , Transfection , Ubiquitination
6.
Cancer Res ; 72(17): 4289-93, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22915759

ABSTRACT

The human CCND1 gene, which encodes the cell-cycle protein cyclin D1, is one of the most frequently amplified genes in human cancers. Cyclin D1 activates the cyclin-dependent kinases CDK4 and CDK6 and drives cell proliferation. Beyond the cell-cycle role, the full repertoire of cyclin D1 functions in cancer cells is still unclear. Emerging evidence indicates that cyclin D1 may play a role in DNA damage response. In this review, we discuss observations linking cyclin D1 to DNA damage repair and summarize our recent findings, which show a cyclin D1 function in homologous recombination-mediated DNA repair.


Subject(s)
Cyclin D1/metabolism , DNA Damage , DNA Repair , Proteomics , Homologous Recombination , Humans , Molecular Targeted Therapy , Neoplasms/metabolism , Neoplasms/therapy , Proteolysis , Proteome
7.
Mol Cell ; 47(3): 396-409, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22704558

ABSTRACT

Completion of DNA replication after replication stress depends on PCNA, which undergoes monoubiquitination to stimulate direct bypass of DNA lesions by specialized DNA polymerases or is polyubiquitinated to promote recombination-dependent DNA synthesis across DNA lesions by template switching mechanisms. Here we report that the ZRANB3 translocase, a SNF2 family member related to the SIOD disorder SMARCAL1 protein, is recruited by polyubiquitinated PCNA to promote fork restart following replication arrest. ZRANB3 depletion in mammalian cells results in an increased frequency of sister chromatid exchange and DNA damage sensitivity after treatment with agents that cause replication stress. Using in vitro biochemical assays, we show that recombinant ZRANB3 remodels DNA structures mimicking stalled replication forks and disassembles recombination intermediates. We therefore propose that ZRANB3 maintains genomic stability at stalled or collapsed replication forks by facilitating fork restart and limiting inappropriate recombination that could occur during template switching events.


Subject(s)
DNA Helicases/metabolism , DNA Replication/physiology , Genomic Instability/physiology , Polyubiquitin/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Stress, Physiological/genetics , Amino Acid Sequence , Cell Line, Tumor , DNA Damage/physiology , DNA Helicases/genetics , Green Fluorescent Proteins/genetics , Humans , Molecular Sequence Data , Osteosarcoma , Protein Binding/physiology , Recombination, Genetic/physiology , Sister Chromatid Exchange/physiology , Ubiquitination/physiology
8.
Nature ; 474(7350): 230-4, 2011 Jun 08.
Article in English | MEDLINE | ID: mdl-21654808

ABSTRACT

Cyclin D1 is a component of the core cell cycle machinery. Abnormally high levels of cyclin D1 are detected in many human cancer types. To elucidate the molecular functions of cyclin D1 in human cancers, we performed a proteomic screen for cyclin D1 protein partners in several types of human tumours. Analyses of cyclin D1 interactors revealed a network of DNA repair proteins, including RAD51, a recombinase that drives the homologous recombination process. We found that cyclin D1 directly binds RAD51, and that cyclin D1-RAD51 interaction is induced by radiation. Like RAD51, cyclin D1 is recruited to DNA damage sites in a BRCA2-dependent fashion. Reduction of cyclin D1 levels in human cancer cells impaired recruitment of RAD51 to damaged DNA, impeded the homologous recombination-mediated DNA repair, and increased sensitivity of cells to radiation in vitro and in vivo. This effect was seen in cancer cells lacking the retinoblastoma protein, which do not require D-cyclins for proliferation. These findings reveal an unexpected function of a core cell cycle protein in DNA repair and suggest that targeting cyclin D1 may be beneficial also in retinoblastoma-negative cancers which are currently thought to be unaffected by cyclin D1 inhibition.


Subject(s)
Cyclin D1/metabolism , DNA Repair , Neoplasms/metabolism , Protein Interaction Mapping , Rad51 Recombinase/metabolism , Animals , Cell Line, Tumor , Comet Assay , Cyclin D1/deficiency , DNA Damage/radiation effects , DNA Repair/radiation effects , HeLa Cells , Humans , Mice , Neoplasms/genetics , Neoplasms/pathology , Protein Binding/radiation effects , Radiation, Ionizing , Recombination, Genetic/genetics , Retinoblastoma Protein/deficiency
9.
Genes Dev ; 25(7): 685-700, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21406551

ABSTRACT

In response to DNA double-strand breaks (DSBs), BRCA1 forms biochemically distinct complexes with certain other DNA damage response proteins. These structures, some of which are required for homologous recombination (HR)-type DSB repair, concentrate at distinct nuclear foci that demarcate sites of genome breakage. Polyubiquitin binding by one of these structures, the RAP80/BRCA1 complex, is required for efficient BRCA1 focal recruitment, but the relationship of this process to the execution of HR has been unclear. We found that this complex actively suppresses otherwise exaggerated, BRCA1-driven HR. By controlling the kinetics by which other BRCA1-interacting proteins that promote HR concentrate together with BRCA1 in nuclear foci, RAP80/BRCA1 complexes suppress excessive DSB end processing, HR-type DSB repair, and overt chromosomal instability. Since chromosomal instability emerges when BRCA1 HR function is either unbridled or absent, active tuning of BRCA1 activity, executed in nuclear foci, is important to genome integrity maintenance.


Subject(s)
BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Carrier Proteins/metabolism , DNA Repair , Nuclear Proteins/metabolism , Radiation, Ionizing , Recombination, Genetic , Carrier Proteins/genetics , Cell Line, Tumor , Chromosomal Instability , Chromosomes/chemistry , Chromosomes/genetics , Chromosomes/metabolism , DNA Breaks, Double-Stranded , DNA-Binding Proteins , HEK293 Cells , HeLa Cells , Histone Chaperones , Humans , Nuclear Proteins/deficiency , Nuclear Proteins/genetics
10.
J Hepatol ; 55(1): 96-102, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21354236

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is frequently a lethal disease and one of the few malignancies that is still increasing in incidence around the world. Better animal models are highly desired to investigate the molecular basis of HCC and to develop novel therapeutic strategies. Alpha-fetoprotein (Afp) gene is expressed in fetal liver, silenced soon after birth, and highly re-expressed in hepatocellular carcinomas (HCC). We aimed to take advantage of the dramatic re-expression of the Afp gene in HCC to develop a hepatocarcinogenesis reporter (HCR) mouse model for dual-modality, longitudinal in vivo imaging of liver tumor development, and progression. METHODS: Knock in mice were established by placing a thymidinekinase (tk)-luciferase (luc) reporter gene cassette under the transcriptional control of the endogenous Afp promoter. DEN, a liver carcinogen, was used to induce liver tumors, which was monitored by both luc-based bioluminescent (BL) and tk-based positron emission tomography (PET) imaging. RESULTS: The expression profile of luc was identical to that of the endogenous Afp gene during development. As early as 2 months after the exposure to DEN, BLI revealed multifocal signals in the liver, long before the appearance of histologically apparent neoplastic lesions. By 6 months, BL and PET dual imaging showed strong signals in malignant HCC. By serendipity, a strong BL signal was also detected in adult testes, a previously unknown site of Afp expression. CONCLUSIONS: The HCR model enables longitudinal monitoring of liver tumor development and progression, providing a powerful tool in developing chemoprevention and therapeutic strategies for HCC.


Subject(s)
Liver Neoplasms, Experimental/etiology , Luciferases/genetics , Thymidine Kinase/genetics , alpha-Fetoproteins/genetics , Animals , Diethylnitrosamine/toxicity , Disease Models, Animal , Disease Progression , Female , Gene Knock-In Techniques , Genes, Reporter , Humans , Liver Neoplasms, Experimental/diagnostic imaging , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Luminescence , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Positron-Emission Tomography , Precancerous Conditions/diagnostic imaging , Precancerous Conditions/etiology , Precancerous Conditions/genetics , Precancerous Conditions/pathology
11.
World J Gastroenterol ; 16(12): 1482-6, 2010 Mar 28.
Article in English | MEDLINE | ID: mdl-20333788

ABSTRACT

AIM: To investigate whether Recql5, a DNA helicase that plays an important role in the maintenance of genome integrity, is a tumor suppressor in the gastrointestinal tract in mice. METHODS: We generated cohorts of both Recql5-proficient and Recql5-deficient Apc(min/+) mice and compared the tumor susceptibility in their gastrointestinal tracts. RESULTS: Recql5 deficiency in Apc(min/+) mice resulted in a significant increase in the tumor incidence in both the colon (P = 0.0162) and the small intestine (P < 0.01). These findings have provided the first genetic evidence for a tumor suppression role of Recql5 in the gastrointestinal tract of mice. Importantly, since mouse Recql5 and human RECQL5 are highly conserved, these findings also suggest that RECQL5 may be a tumor suppressor for human colon cancer. CONCLUSION: Recql5 has a tumor suppression role in the mouse gastrointestinal tract.


Subject(s)
Adenoma/genetics , Genes, APC , Intestinal Neoplasms/genetics , RecQ Helicases/deficiency , Tumor Suppressor Proteins/deficiency , Adenoma/enzymology , Adenoma/pathology , Adenoma/prevention & control , Animals , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Duodenal Neoplasms/enzymology , Duodenal Neoplasms/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Ileal Neoplasms/enzymology , Ileal Neoplasms/genetics , Intestinal Neoplasms/enzymology , Intestinal Neoplasms/pathology , Intestinal Neoplasms/prevention & control , Jejunal Neoplasms/enzymology , Jejunal Neoplasms/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RecQ Helicases/genetics , Tumor Suppressor Proteins/genetics
12.
Cancer Treat Rev ; 35(7): 590-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19635647

ABSTRACT

Increased chemo-resistance and radio-resistance of cancer cells is a major obstacle in the treatment and management of malignant cancers. An important mechanism that underlies the development of such therapeutic resistance is that cancer cells recognize DNA lesions induced by DNA-damaging agents and by ionizing radiation, and repair these lesions by activating various DNA repair pathways. Therefore, Use of pharmacological agents that can inhibit certain DNA repair pathways in cancer cells has the potential for enhancing the targeted cytotoxicity of anticancer treatments and reversing the associated therapeutic resistance associated with DNA repair; such agents, offering a promising opportunity to achieve better therapeutic efficacy. Here we review the major DNA repair pathways and discuss recent advances in the development of novel inhibitors of DNA repair pathways; many of these agents are under preclinical/clinical investigation.


Subject(s)
DNA Repair/drug effects , Neoplasms/drug therapy , Neoplasms/genetics , DNA Damage , Drug Resistance, Neoplasm , Humans
13.
Mol Biol Cell ; 20(1): 114-23, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18987339

ABSTRACT

Disruption of replication can lead to loss of genome integrity and increase of cancer susceptibility in mammals. Thus, a replication impediment constitutes a formidable challenge to these organisms. Recent studies indicate that homologous recombination (HR) plays an important role in suppressing genome instability and promoting cell survival after exposure to various replication inhibitors, including a topoisomerase I inhibitor, camptothecin (CPT). Here, we report that the deletion of RecQ helicase Recql5 in mouse ES cells and embryonic fibroblast (MEF) cells resulted in a significant increase in CPT sensitivity and a profound reduction in DNA replication after the treatment with CPT, but not other DNA-damaging agents. This CPT-induced cell death is replication dependent and occurs primarily after the cells had exited the first cell cycle after CPT treatment. Furthermore, we show that Recql5 functions nonredundantly with Rad51, a key factor for HR to protect mouse ES cells from CPT-induced cytotoxicity. These new findings strongly suggest that Recql5 plays an important role in maintaining active DNA replication to prevent the collapse of replication forks and the accumulation of DSBs in order to preserve genome integrity and to prevent cell death after replication stress as a result of topoisomerase I poisoning.


Subject(s)
Camptothecin/pharmacology , Cell Survival/drug effects , DNA Replication , Embryonic Stem Cells , Enzyme Inhibitors/pharmacology , Fibroblasts , RecQ Helicases/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/physiology , Cell Survival/physiology , Cells, Cultured , DNA Replication/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , Mice , Rad51 Recombinase/metabolism , RecQ Helicases/genetics
14.
Genes Dev ; 21(23): 3073-84, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18003859

ABSTRACT

Members of the RecQ helicase family play critical roles in genome maintenance. There are five RecQ homologs in mammals, and defects in three of these (BLM, WRN, and RECQL4) give rise to cancer predisposition syndromes in humans. RECQL and RECQL5 have not been associated with a human disease. Here we show that deletion of Recql5 in mice results in cancer susceptibility. Recql5-deficient cells exhibit elevated frequencies of spontaneous DNA double-strand breaks and homologous recombination (HR) as scored using a reporter that harbors a direct repeat, and are prone to gross chromosomal rearrangements in response to replication stress. To understand how RECQL5 regulates HR, we use purified proteins to demonstrate that human RECQL5 binds the Rad51 recombinase and inhibits Rad51-mediated D-loop formation. By biochemical means and electron microscopy, we show that RECQL5 displaces Rad51 from single-stranded DNA (ssDNA) in a reaction that requires ATP hydrolysis and RPA. Together, our results identify RECQL5 as an important tumor suppressor that may act by preventing inappropriate HR events via Rad51 presynaptic filament disruption.


Subject(s)
Neoplasms, Experimental/prevention & control , Rad51 Recombinase/metabolism , RecQ Helicases/metabolism , Recombination, Genetic , Adenosine Triphosphate/metabolism , Animals , Cell Line , DNA Breaks, Double-Stranded , Genomic Instability , Histones/metabolism , Humans , Hydrolysis , Loss of Heterozygosity , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Mutation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Rad51 Recombinase/genetics , RecQ Helicases/deficiency , RecQ Helicases/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
Mol Cell Biol ; 25(9): 3431-42, 2005 May.
Article in English | MEDLINE | ID: mdl-15831450

ABSTRACT

In eukaryotes, crossovers in mitotic cells can have deleterious consequences and therefore must be suppressed. Mutations in BLM give rise to Bloom syndrome, a disease that is characterized by an elevated rate of crossovers and increased cancer susceptibility. However, simple eukaryotes such as Saccharomyces cerevisiae have multiple pathways for suppressing crossovers, suggesting that mammals also have multiple pathways for controlling crossovers in their mitotic cells. We show here that in mouse embryonic stem (ES) cells, mutations in either the Bloom syndrome homologue (Blm) or the Recql5 genes result in a significant increase in the frequency of sister chromatid exchange (SCE), whereas deleting both Blm and Recql5 lead to an even higher frequency of SCE. These data indicate that Blm and Recql5 have nonredundant roles in suppressing crossovers in mouse ES cells. Furthermore, we show that mouse embryonic fibroblasts derived from Recql5 knockout mice also exhibit a significantly increased frequency of SCE compared with the corresponding wild-type control. Thus, this study identifies a previously unknown Recql5-dependent, Blm-independent pathway for suppressing crossovers during mitosis in mice.


Subject(s)
Adenosine Triphosphatases/physiology , Bloom Syndrome/genetics , Crossing Over, Genetic/genetics , DNA Helicases/physiology , Sister Chromatid Exchange/genetics , Adenosine Triphosphatases/genetics , Animals , Crossing Over, Genetic/physiology , Crossing Over, Genetic/radiation effects , DNA Helicases/genetics , Fibroblasts/radiation effects , Gamma Rays , Gene Deletion , Gene Silencing , Gene Targeting , Mice , Mice, Knockout , Mitosis/genetics , Mitosis/physiology , Mitosis/radiation effects , RecQ Helicases , Sister Chromatid Exchange/physiology , Stem Cells/radiation effects
16.
Lin Chuang Er Bi Yan Hou Ke Za Zhi ; 16(5): 196-7, 2002 May.
Article in Chinese | MEDLINE | ID: mdl-12592648

ABSTRACT

OBJECTIVE: To study the treatment of inverted papilloma (NIP) with intracranial extension. METHOD: Nine patients of NIP all suffered from the operation. Six patients with anterior skull base erosion suffered from large tumor resections via combined craniofacial and cranial approach. Three patients suffered from small tumor resections via transfrontal and transethmoidal sinus approach. The defects of basicranial bone and dura were repaired at the same time. Five patients were found the tissue canceration in the postoperation. RESULT: In 7 patients followed-up from 3 to 13 years, no one has recurrenced. CONCLUSION: The canceration probability of NIP with intracranial extension should be sufficiently considered. The tumor resection via combined craniofacial approach or transfrontal approach must be based on the tumor size and extension, and removed completely.


Subject(s)
Nose Neoplasms/surgery , Papilloma, Inverted/surgery , Paranasal Sinus Neoplasms/surgery , Adult , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Invasiveness , Nose Neoplasms/pathology , Papilloma, Inverted/pathology , Paranasal Sinus Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...