Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 15 de 15
Filtrer
2.
Mol Ther Methods Clin Dev ; 31: 101135, 2023 Dec 14.
Article de Anglais | MEDLINE | ID: mdl-38027064

RÉSUMÉ

Immunotherapy of acute myeloid leukemia (AML) has been challenging because the lack of tumor-specific antigens results in "on-target, off-tumor" toxicity. To unlock the full potential of AML therapies, we used CRISPR-Cas9 to genetically ablate the myeloid protein CD33 from healthy donor hematopoietic stem and progenitor cells (HSPCs), creating tremtelectogene empogeditemcel (trem-cel). Trem-cel is a HSPC transplant product designed to provide a reconstituted hematopoietic compartment that is resistant to anti-CD33 drug cytotoxicity. Here, we describe preclinical studies and process development of clinical-scale manufacturing of trem-cel. Preclinical data showed proof-of-concept with loss of CD33 surface protein and no impact on myeloid cell differentiation or function. At clinical scale, trem-cel could be manufactured reproducibly, routinely achieving >70% CD33 editing with no effect on cell viability, differentiation, and function. Trem-cel pharmacology studies using mouse xenograft models showed long-term engraftment, multilineage differentiation, and persistence of gene editing. Toxicology assessment revealed no adverse findings, and no significant or reproducible off-target editing events. Importantly, CD33-knockout myeloid cells were resistant to the CD33-targeted agent gemtuzumab ozogamicin in vitro and in vivo. These studies supported the initiation of the first-in-human, multicenter clinical trial evaluating the safety and efficacy of trem-cel in patients with AML (NCT04849910).

3.
Oncogene ; 40(38): 5718-5729, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34331013

RÉSUMÉ

Melanomas driven by loss of the NF1 tumor suppressor have a high risk of treatment failure and effective therapies have not been developed. Here we show that loss-of-function mutations of nf1 and pten result in aggressive melanomas in zebrafish, representing the first animal model of NF1-mutant melanomas harboring PTEN loss. MEK or PI3K inhibitors show little activity when given alone due to cross-talk between the pathways, and high toxicity when given together. The mTOR inhibitors, sirolimus, everolimus, and temsirolimus, were the most active single agents tested, potently induced tumor-suppressive autophagy, but not apoptosis. Because addition of the BCL2 inhibitor venetoclax resulted in compensatory upregulation of MCL1, we established a three-drug combination composed of sirolimus, venetoclax, and the MCL1 inhibitor S63845. This well-tolerated drug combination potently and synergistically induces apoptosis in both zebrafish and human NF1/PTEN-deficient melanoma cells, providing preclinical evidence justifying an early-stage clinical trial in patients with NF1/PTEN-deficient melanoma.


Sujet(s)
Composés hétérocycliques bicycliques/administration et posologie , Inhibiteurs de mTOR/administration et posologie , Mélanome/traitement médicamenteux , Neurofibromine-1/génétique , Phosphohydrolase PTEN/génétique , Pyrimidines/administration et posologie , Sulfonamides/administration et posologie , Thiophènes/administration et posologie , Animaux , Composés hétérocycliques bicycliques/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Évérolimus/administration et posologie , Évérolimus/pharmacologie , Humains , Mutation perte de fonction , Inhibiteurs de mTOR/pharmacologie , Mélanome/génétique , Mélanome/anatomopathologie , Protéine Mcl-1/antagonistes et inhibiteurs , Protéines proto-oncogènes c-bcl-2/antagonistes et inhibiteurs , Pyrimidines/pharmacologie , Sirolimus/administration et posologie , Sirolimus/analogues et dérivés , Sirolimus/pharmacologie , Sulfonamides/pharmacologie , Thiophènes/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Danio zébré
5.
Br J Haematol ; 177(2): 271-282, 2017 04.
Article de Anglais | MEDLINE | ID: mdl-28295194

RÉSUMÉ

Activation of tyrosine kinase 2 (TYK2) contributes to the aberrant survival of T-cell acute lymphoblastic leukaemia (T-ALL) cells. Here we demonstrate the anti-leukaemic activity of a novel TYK2 inhibitor, NDI-031301. NDI-031301 is a potent and selective inhibitor of TYK2 that induced robust growth inhibition of human T-ALL cell lines. NDI-031301 treatment of human T-ALL cell lines resulted in induction of apoptosis that was not observed with the JAK inhibitors tofacitinib and baricitinib. Further investigation revealed that NDI-031301 treatment uniquely leads to activation of three mitogen-activated protein kinases (MAPKs), resulting in phosphorylation of ERK, SAPK/JNK and p38 MAPK coincident with PARP cleavage. Activation of p38 MAPK occurred within 1 h of NDI-031301 treatment and was responsible for NDI-031301-induced T-ALL cell death, as pharmacological inhibition of p38 MAPK partially rescued apoptosis induced by TYK2 inhibitor. Finally, daily oral administration of NDI-031301 at 100 mg/kg bid to immunodeficient mice engrafted with KOPT-K1 T-ALL cells was well tolerated, and led to decreased tumour burden and a significant survival benefit. These results support selective inhibition of TYK2 as a promising potential therapeutic strategy for T-ALL.


Sujet(s)
Leucémie-lymphome lymphoblastique à précurseurs T/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , TYK2 Kinase/antagonistes et inhibiteurs , Animaux , Lignée cellulaire tumorale , Femelle , Humains , Souris , Souris de lignée NOD , Souris SCID , Leucémie-lymphome lymphoblastique à précurseurs T/enzymologie , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Nat Commun ; 8: 14385, 2017 02 09.
Article de Anglais | MEDLINE | ID: mdl-28181482

RÉSUMÉ

The non-coding regions of tumour cell genomes harbour a considerable fraction of total DNA sequence variation, but the functional contribution of these variants to tumorigenesis is ill-defined. Among these non-coding variants, somatic insertions are among the least well characterized due to challenges with interpreting short-read DNA sequences. Here, using a combination of Chip-seq to enrich enhancer DNA and a computational approach with multiple DNA alignment procedures, we identify enhancer-associated small insertion variants. Among the 102 tumour cell genomes we analyse, small insertions are frequently observed in enhancer DNA sequences near known oncogenes. Further study of one insertion, somatically acquired in primary leukaemia tumour genomes, reveals that it nucleates formation of an active enhancer that drives expression of the LMO2 oncogene. The approach described here to identify enhancer-associated small insertion variants provides a foundation for further study of these abnormalities across human cancers.


Sujet(s)
Éléments activateurs (génétique) , Génome humain , Mutagenèse par insertion/génétique , Oncogènes , Adolescent , Adulte , Séquence nucléotidique , Lignée cellulaire tumorale , Enfant , Enfant d'âge préscolaire , Régulation de l'expression des gènes dans la leucémie , Humains , Nourrisson , Leucémie-lymphome à cellules T de l'adulte/génétique , Reproductibilité des résultats , Jeune adulte
8.
Cancer Cell ; 29(4): 574-586, 2016 04 11.
Article de Anglais | MEDLINE | ID: mdl-27070704

RÉSUMÉ

More than 90% of drugs with preclinical activity fail in human trials, largely due to insufficient efficacy. We hypothesized that adequately powered trials of patient-derived xenografts (PDX) in mice could efficiently define therapeutic activity across heterogeneous tumors. To address this hypothesis, we established a large, publicly available repository of well-characterized leukemia and lymphoma PDXs that undergo orthotopic engraftment, called the Public Repository of Xenografts (PRoXe). PRoXe includes all de-identified information relevant to the primary specimens and the PDXs derived from them. Using this repository, we demonstrate that large studies of acute leukemia PDXs that mimic human randomized clinical trials can characterize drug efficacy and generate transcriptional, functional, and proteomic biomarkers in both treatment-naive and relapsed/refractory disease.


Sujet(s)
Hétérogreffes , Leucémies/anatomopathologie , Lymphomes/anatomopathologie , Banques de tissus , Tests d'activité antitumorale sur modèle de xénogreffe , Animaux , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Marqueurs biologiques tumoraux , Lignage cellulaire , Femelle , Analyse de profil d'expression de gènes , Gènes p53 , Humains , Internet , Isoquinoléines/pharmacologie , Isoquinoléines/usage thérapeutique , Leucémies/métabolisme , Leucémie expérimentale/traitement médicamenteux , Lymphomes/métabolisme , Mâle , Souris , Souris de lignée NOD , Thérapie moléculaire ciblée , Protéines tumorales/antagonistes et inhibiteurs , Transplantation tumorale , Phénotype , Pipérazines/pharmacologie , Pipérazines/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie , Protéome , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Répartition aléatoire , Essais contrôlés randomisés comme sujet/méthodes , Plan de recherche , Transcriptome
9.
Science ; 346(6215): 1373-7, 2014 Dec 12.
Article de Anglais | MEDLINE | ID: mdl-25394790

RÉSUMÉ

In certain human cancers, the expression of critical oncogenes is driven from large regulatory elements, called super-enhancers, that recruit much of the cell's transcriptional apparatus and are defined by extensive acetylation of histone H3 lysine 27 (H3K27ac). In a subset of T-cell acute lymphoblastic leukemia (T-ALL) cases, we found that heterozygous somatic mutations are acquired that introduce binding motifs for the MYB transcription factor in a precise noncoding site, which creates a super-enhancer upstream of the TAL1 oncogene. MYB binds to this new site and recruits its H3K27 acetylase-binding partner CBP, as well as core components of a major leukemogenic transcriptional complex that contains RUNX1, GATA-3, and TAL1 itself. Additionally, most endogenous super-enhancers found in T-ALL cells are occupied by MYB and CBP, which suggests a general role for MYB in super-enhancer initiation. Thus, this study identifies a genetic mechanism responsible for the generation of oncogenic super-enhancers in malignant cells.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , ADN intergénique , Éléments activateurs (génétique) , Régulation de l'expression des gènes tumoraux , Mutation de type INDEL , Mutation , Leucémie-lymphome lymphoblastique à précurseurs T/génétique , Protéines proto-oncogènes/génétique , Acétylation , Séquence nucléotidique , Sites de fixation , Lignée cellulaire tumorale , Histone/métabolisme , Humains , Données de séquences moléculaires , Oncogènes , Motifs et domaines d'intéraction protéique , Protéines proto-oncogènes c-myb/métabolisme , Protéine-1 de la lleucémie lymphoïde aiguë à cellules T
10.
Gastroenterology ; 147(4): 882-892.e8, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-24998203

RÉSUMÉ

BACKGROUND & AIMS: Development of pancreatic ductal adenocarcinoma (PDAC) involves activation of c-Ki-ras2 Kirsten rat sarcoma oncogene homolog (KRAS) signaling, but little is known about the roles of proteins that regulate the activity of oncogenic KRAS. We investigated the activities of proteins that interact with KRAS in PDAC cells. METHODS: We used mass spectrometry to demonstrate that heterogeneous nuclear ribonucleoproteins (HNRNP) A2 and B1 (encoded by the gene HNRNPA2B1) interact with KRAS G12V. We used co-immunoprecipitation analyses to study interactions between HNRNPA2B1 and KRAS in KRAS-dependent and KRAS-independent PDAC cell lines. We knocked down HNRNPA2B1 using small hairpin RNAs and measured viability, anchorage-independent proliferation, and growth of xenograft tumors in mice. We studied KRAS phosphorylation using the Phos-tag system. RESULTS: We found that interactions between HRNPA2B1 and KRAS correlated with KRAS-dependency of some human PDAC cell lines. Knock down of HNRNPA2B1 significantly reduced viability, anchorage-independent proliferation, and formation of xenograft tumors by KRAS-dependent PDAC cells. HNRNPA2B1 knock down also increased apoptosis of KRAS-dependent PDAC cells, inactivated c-akt murine thymoma oncogene homolog 1 signaling via mammalian target of rapamycin, and reduced interaction between KRAS and phosphatidylinositide 3-kinase. Interaction between HNRNPA2B1 and KRAS required KRAS phosphorylation at serine 181. CONCLUSIONS: In KRAS-dependent PDAC cell lines, HNRNPA2B1 interacts with and regulates the activity of KRAS G12V and G12D. HNRNPA2B1 is required for KRAS activation of c-akt murine thymoma oncogene homolog 1-mammalian target of rapamycin signaling, interaction with phosphatidylinositide 3-kinase, and PDAC cell survival and tumor formation in mice. HNRNPA2B1 might be a target for treatment of pancreatic cancer.


Sujet(s)
Carcinome du canal pancréatique/métabolisme , Ribonucléoprotéine nucléaire hétérogène du groupe A-B/métabolisme , Tumeurs du pancréas/métabolisme , Protéines proto-oncogènes/métabolisme , Protéines G ras/métabolisme , Animaux , Apoptose , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Adhérence cellulaire , Prolifération cellulaire , Survie cellulaire , Régulation de l'expression des gènes tumoraux , Techniques de knock-down de gènes , Cellules HeLa , Ribonucléoprotéine nucléaire hétérogène du groupe A-B/génétique , Humains , Souris , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Phosphatidylinositol 3-kinase/métabolisme , Phosphorylation , Liaison aux protéines , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes p21(ras) , Interférence par ARN , Transduction du signal , Sérine-thréonine kinases TOR/métabolisme , Facteurs temps , Transfection , Charge tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Protéines G ras/génétique
11.
Br J Haematol ; 161(1): 117-27, 2013 Apr.
Article de Anglais | MEDLINE | ID: mdl-23373539

RÉSUMÉ

This study explored the anti-leukaemic efficacy of novel irreversible inhibitors of the major nuclear export receptor, chromosome region maintenance 1 (CRM1, also termed XPO1). We found that these novel CRM1 antagonists, termed SINE (Selective Inhibitors of Nuclear Export), induced rapid apoptosis at low nanomolar concentrations in a panel of 14 human T-cell acute lymphoblastic leukaemia (T-ALL) cell lines representing different molecular subtypes of the disease. To assess in vivo anti-leukaemia cell activity, we engrafted immunodeficient mice intravenously with the human T-ALL MOLT-4 cells, which harbour activating mutations of NOTCH1 and NRAS as well as loss of function of the CDKN2A, PTEN and TP53 tumour suppressors and express a high level of oncogenic transcription factor TAL1. Importantly, we examined the in vivo anti-leukaemic efficacy of the clinical SINE compound KPT-330 against T-ALL and acute myeloid leukaemia (AML) cells. These studies demonstrated striking in vivo activity of KPT-330 against T-ALL and AML cells, with little toxicity to normal murine haematopoietic cells. Taken together, our results show that SINE CRM1 antagonists represent promising 'first-in-class' drugs with a novel mechanism of action and wide therapeutic index, and imply that drugs of this class show promise for the targeted therapy of T-ALL and AML.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Caryophérines/antagonistes et inhibiteurs , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs T/traitement médicamenteux , Récepteurs cytoplasmiques et nucléaires/antagonistes et inhibiteurs , Transport nucléaire actif/effets des médicaments et des substances chimiques , Animaux , Antinéoplasiques/administration et posologie , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Cellules sanguines/effets des médicaments et des substances chimiques , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Noyau de la cellule/métabolisme , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Femelle , Humains , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Souris , Souris de lignée NOD , Souris SCID , Protéines nucléaires/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs T/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs T/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe ,
12.
Methods Cell Biol ; 105: 309-37, 2011.
Article de Anglais | MEDLINE | ID: mdl-21951536

RÉSUMÉ

Zebrafish provide an exciting animal model system for the study of human cancers. During the last few years many zebrafish models of cancer have been generated that recapitulate human hematologic malignancies and solid tumors. Concurrent technological advances have significantly improved the genetic tractability and unique advantage of in vivo imaging in zebrafish, providing a means to dissect the molecular pathways underlying tumor initiation, progression and metastasis. Comparisons of cancer-associated gene expression profiles have demonstrated a high degree of similarity in the gene signatures of specific types of tumor cells in fish and humans, indicating that the contributing genetic pathways leading to cancer are evolutionarily conserved. Furthermore, the high fecundity, optical clarity and small embryo size of zebrafish continue to make it particularly amenable to performing whole-organism small molecule screens to identify targets for therapeutic development. This chapter reviews a wide array of these zebrafish cancer models and illustrates the advantages of the zebrafish system for exploring the molecular mechanisms governing cancer-related cellular processes.


Sujet(s)
Transformation cellulaire néoplasique/génétique , Techniques de transfert de gènes , Tests de criblage à haut débit , Oncologie médicale/méthodes , Mutagenèse/génétique , Métastase tumorale , Protéines suppresseurs de tumeurs/déficit , Danio zébré/génétique , Animaux , Transformation cellulaire néoplasique/métabolisme , Modèles animaux de maladie humaine , Découverte de médicament , Embryon non mammalien , Analyse de profil d'expression de gènes , Humains , Leucémies/embryologie , Leucémies/génétique , Leucémies/anatomopathologie , Mélanocytes/métabolisme , Mélanocytes/anatomopathologie , Mélanome/embryologie , Mélanome/génétique , Mélanome/anatomopathologie , Métastase tumorale/génétique , Tumeurs du pancréas/embryologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Rhabdomyosarcome/embryologie , Rhabdomyosarcome/génétique , Rhabdomyosarcome/anatomopathologie , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/pharmacologie , Protéines suppresseurs de tumeurs/génétique , Danio zébré/embryologie , Danio zébré/métabolisme
13.
Nat Struct Mol Biol ; 17(10): 1255-9, 2010 Oct.
Article de Anglais | MEDLINE | ID: mdl-20871616

RÉSUMÉ

Homologous recombination mediated by RAD51 recombinase helps eliminate chromosomal lesions, such as DNA double-strand breaks induced by radiation or arising from injured DNA replication forks. The tumor suppressors BRCA2 and PALB2 act together to deliver RAD51 to chromosomal lesions to initiate repair. Here we document a new function of PALB2: to enhance RAD51's ability to form the D loop. We show that PALB2 binds DNA and physically interacts with RAD51. Notably, although PALB2 alone stimulates D-loop formation, it has a cooperative effect with RAD51AP1, an enhancer of RAD51. This stimulation stems from the ability of PALB2 to function with RAD51 and RAD51AP1 to assemble the synaptic complex. Our results demonstrate the multifaceted role of PALB2 in chromosome damage repair. Because PALB2 mutations can cause cancer or Fanconi anemia, our findings shed light on the mechanism of tumor suppression in humans.


Sujet(s)
Protéine BRCA2/physiologie , Tumeurs du sein/métabolisme , Réparation de l'ADN/physiologie , ADN tumoral/métabolisme , Protéines de liaison à l'ADN/physiologie , Protéines tumorales/physiologie , Protéines nucléaires/physiologie , Rad51 Recombinase/physiologie , Recombinaison génétique/physiologie , Protéines suppresseurs de tumeurs/physiologie , Protéines régulatrices de l'apoptose , Protéine BRCA2/composition chimique , Protéines de liaison à l'ADN/composition chimique , Protéine du groupe de complémentation N de l'anémie de Fanconi , Femelle , Humains , Complexes multiprotéiques , Protéines tumorales/composition chimique , Protéines nucléaires/composition chimique , Protéines nucléaires/génétique , Fragments peptidiques/métabolisme , Liaison aux protéines , Cartographie d'interactions entre protéines , Protéines de liaison à l'ARN , Rad51 Recombinase/composition chimique , Protéines de fusion recombinantes/composition chimique , Protéines de fusion recombinantes/physiologie , Protéines suppresseurs de tumeurs/composition chimique , Protéines suppresseurs de tumeurs/génétique
14.
Cancer Res ; 67(3): 1254-61, 2007 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-17283162

RÉSUMÉ

The transcription factor signal transducer and activator of transcription (STAT) 1 can mediate antiproliferative and proapoptotic effects in cancer cells, and a number of mechanisms have been found whereby STAT1 signaling is attenuated in tumors thereby increasing their malignant behavior. Thus, enhancing gene transcription mediated by STAT1 may be an effective approach to cancer therapy. A high-throughput screen was developed to identify molecules that could enhance STAT1-dependent gene expression. Through this approach, it was found that 2-(1,8-naphthyridin-2-yl)phenol (2-NP) caused a 2-fold increase in STAT1-dependent reporter gene expression compared with that seen with maximally effective concentrations of IFN-gamma alone. This effect was specific to STAT1 because 2-NP had no effect on unrelated transcription factors such as nuclear factor (NF) kappaB or the highly homologous transcription factor STAT3. STAT1-dependent gene activation was enhanced by this compound in a variety of human and murine cell lines and was independent of the stimulus used. Furthermore, 2-NP enhanced the expression of the bona fide endogenous STAT1 target gene interferon regulatory factor 1. 2-NP increased the duration of STAT1 tyrosine phosphorylation in response to IFN-gamma, and this may underlie its enhancement of STAT1-dependent transcription. Reflecting the fact that STAT1 can exert tumor-suppressive effects, 2-NP enhanced the ability of IFN-gamma to inhibit the proliferation of human breast cancer and fibrosarcoma cells. Tumor cells lacking STAT1 were unaffected by either IFN-gamma or 2-NP. These findings indicate that enhancement of STAT1 transcriptional activity may have utility in anticancer therapies, and that cell-based screens for modulators of transcription factor function can be a useful approach for drug discovery.


Sujet(s)
Interféron gamma/pharmacologie , Facteur de transcription STAT-1/biosynthèse , Transcription génétique/effets des médicaments et des substances chimiques , Animaux , Processus de croissance cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Synergie des médicaments , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Souris , Cellules NIH 3T3 , Naphtyridines , Phosphorylation/effets des médicaments et des substances chimiques , Facteur de transcription STAT-1/génétique , Facteur de transcription STAT-1/métabolisme , Activation de la transcription
15.
J Biol Chem ; 281(17): 11649-57, 2006 Apr 28.
Article de Anglais | MEDLINE | ID: mdl-16513631

RÉSUMÉ

BRCA2 likely exerts its tumor suppressor function by enhancing the efficiency of the homology-directed repair of injured chromosomes. To help define the DNA repair role of BRCA2, we expressed and purified a polypeptide, BRC3/4-DBD, that harbors its BRC3 and BRC4 repeats and DNA binding domain. BRC3/4-DBD interacted with hRad51 and bound DNA with a distinct preference for single-stranded (ss) DNA. Importantly we demonstrated by biochemical means and electron microscopy that BRC3/4-DBD nucleates hRad51 onto ssDNA and acts as a recombination mediator in enabling hRad51 to utilize replication protein A-coated ssDNA as recombination substrate. These functions of BRC3/4-DBD required both the BRC repeats and the BRCA2 DNA binding domain. The results thus clarify the role of BRCA2 in Rad51-dependent DNA recombination and repair, and the experimental strategies described herein should be valuable for systematically deciphering this BRCA2 function.


Sujet(s)
Protéine BRCA2/métabolisme , Réparation de l'ADN , ADN simple brin/métabolisme , Fragments peptidiques/métabolisme , Rad51 Recombinase/génétique , Recombinaison génétique , Protéine BRCA2/génétique , Altération de l'ADN , Amorces ADN/composition chimique , ADN simple brin/génétique , Humains , Fragments peptidiques/génétique , Rad51 Recombinase/métabolisme , Protéine A de réplication/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE