Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
bioRxiv ; 2023 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-37720033

RESUMO

Deficiency for the repair of DNA double-strand breaks (DSBs) via homologous recombination (HR) leads to chromosomal instability and diseases such as cancer. Yet, defective HR also results in vulnerabilities that can be exploited for targeted therapy. Here, we identify such a vulnerability and show that BRCA1-deficient cells are dependent on the long-range end-resection factor EXO1 for survival. EXO1 loss results in DNA replication-induced lesions decorated by poly(ADP-ribose)-chains. In cells that lack both BRCA1 and EXO1, this is accompanied by unresolved DSBs due to impaired single-strand annealing (SSA), a DSB repair process that requires the activity of both proteins. In contrast, BRCA2-deficient cells have increased SSA, also in the absence of EXO1, and hence are not dependent on EXO1 for survival. In agreement with our mechanistic data, BRCA1-mutated tumours have elevated EXO1 expression and contain more genomic signatures of SSA compared to BRCA1-proficient tumours. Collectively, our data indicate that EXO1 is a promising novel target for treatment of BRCA1-deficient tumours.

2.
Sci Rep ; 13(1): 10720, 2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-37400554

RESUMO

Differences in the clinical course and treatment responses in individual patients with advanced renal cell carcinoma (RCC) can largely be explained by the different genomics of this disease. To improve the personalized treatment strategy and survival outcomes for patients with advanced RCC, the genomic make-up in patients with advanced RCC was investigated to identify putative actionable variants and signatures. In this prospective multicenter study (NCT01855477), whole-genome sequencing (WGS) data of locally advanced and metastatic tissue biopsies and matched whole-blood samples were collected from 91 patients with histopathologically confirmed RCC. WGS data were analyzed for small somatic variants, copy-number alterations and structural variants. For a subgroup of patients, RNA sequencing (RNA-Seq) data could be analyzed. RNA-Seq data were clustered on immunogenic and angiogenic gene expression patterns according to a previously developed angio-immunogenic gene signature. In all patients with papillary and clear cell RCC, putative actionable drug targets were detected by WGS, of which 94% were on-label available. RNA-Seq data of clear cell and papillary RCC were clustered using a previously developed angio-immunogenic gene signature. Analyses of driver mutations and RNA-Seq data revealed clear differences among different RCC subtypes, showing the added value of WGS and RNA-Seq over clinicopathological data. By improving both histological subtyping and the selection of treatment according to actionable targets and immune signatures, WGS and RNA-Seq may improve therapeutic decision making for most patients with advanced RCC, including patients with non-clear cell RCC for whom no standard treatment is available to data. Prospective clinical trials are needed to evaluate the impact of genomic and transcriptomic diagnostics on survival outcome for advanced RCC patients.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Humanos , Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , Transcriptoma , Estudos Prospectivos , Genômica
3.
ESMO Open ; 7(6): 100611, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36463731

RESUMO

BACKGROUND: In ∼3%-5% of patients with metastatic disease, tumor origin remains unknown despite modern imaging techniques and extensive pathology work-up. With long diagnostic delays and limited and ineffective therapy options, the clinical outcome of patients with cancer of unknown primary (CUP) remains poor. Large-scale genome sequencing studies have revealed that tumor types can be predicted based on distinct patterns of somatic variants and other genomic characteristics. Moreover, actionable genomic events are present in almost half of CUP patients. This study investigated the clinical value of whole genome sequencing (WGS) in terms of primary tumor identification and detection of actionable events, in the routine diagnostic work-up of CUP patients. PATIENTS AND METHODS: A WGS-based tumor type 'cancer of unknown primary prediction algorithm' (CUPPA) was developed based on previously described principles and validated on a large pan-cancer WGS database of metastatic cancer patients (>4000 samples) and 254 independent patients, respectively. We assessed the clinical value of this prediction algorithm as part of routine WGS-based diagnostic work-up for 72 CUP patients. RESULTS: CUPPA correctly predicted the primary tumor type in 78% of samples in the independent validation cohort (194/254 patients). High-confidence predictions (>95% precision) were obtained for 162/254 patients (64%). When integrated in the diagnostic work-up of CUP patients, CUPPA could identify a primary tumor type for 49/72 patients (68%). Most common diagnoses included non-small-cell lung (n = 7), gastroesophageal (n = 4), pancreatic (n = 4), and colorectal cancer (n = 3). Actionable events with matched therapy options in clinical trials were identified in 47% of patients. CONCLUSIONS: Genome-based tumor type prediction can predict cancer diagnoses with high accuracy when integrated in the routine diagnostic work-up of patients with metastatic cancer. With identification of the primary tumor type in the majority of patients and detection of actionable events, WGS is a valuable diagnostic tool for patients with CUP.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Neoplasias Primárias Desconhecidas , Humanos , Neoplasias Primárias Desconhecidas/diagnóstico , Neoplasias Primárias Desconhecidas/genética , Neoplasias Primárias Desconhecidas/tratamento farmacológico , Genômica , Sequenciamento Completo do Genoma
4.
ESMO Open ; 6(3): 100103, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33887686

RESUMO

BACKGROUND: Organoid technology has recently emerged as a powerful tool to assess drug sensitivity of individual patient tumors in vitro. Organoids may therefore represent a new avenue for precision medicine, as this circumvents many of the complexities associated with DNA- or transcriptional-profiling. MATERIALS AND METHODS: The SENSOR trial was a single-arm, single-center, prospective intervention trial to evaluate the feasibility of patient-derived organoids to allocate patients for treatment with off-label or investigational agents. The primary endpoint was an objective response rate of ≥20%. Patients underwent a biopsy for culture before commencing their last round standard of care. Organoids were exposed to a panel of eight drugs and patients were treated after progression on standard-of-care treatment and when a clear signal of antitumor activity was identified in vitro. RESULTS: Sixty-one patients were included and we generated 31 organoids of 54 eligible patients. Twenty-five cultures were subjected to drug screening and 19 organoids exhibited substantial responses to one or more drugs. Three patients underwent treatment with vistusertib and three with capivasertib. Despite drug sensitivity of organoids, patients did not demonstrate objective clinical responses to the recommended treatment. CONCLUSIONS: Organoid technology had limited value as a tool for precision medicine in this patient population because a large fraction of patients could not undergo treatment or because the recommended treatment did not elicit an objective response. We identified several essential parameters, such as the culture success rate, clinical deterioration of patients during standard of care, and rational design of drug panels that need to be accounted for in organoid-guided clinical studies.


Assuntos
Neoplasias Colorretais , Preparações Farmacêuticas , Neoplasias Colorretais/tratamento farmacológico , Humanos , Organoides , Medicina de Precisão , Estudos Prospectivos
5.
Nature ; 574(7776): 127-131, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31570881

RESUMO

The large-scale genetic profiling of tumours can identify potentially actionable molecular variants for which approved anticancer drugs are available1-3. However, when patients with such variants are treated with drugs outside of their approved label, successes and failures of targeted therapy are not systematically collected or shared. We therefore initiated the Drug Rediscovery protocol, an adaptive, precision-oncology trial that aims to identify signals of activity in cohorts of patients, with defined tumour types and molecular variants, who are being treated with anticancer drugs outside of their approved label. To be eligible for the trial, patients have to have exhausted or declined standard therapies, and have malignancies with potentially actionable variants for which no approved anticancer drugs are available. Here we show an overall rate of clinical benefit-defined as complete or partial response, or as stable disease beyond 16 weeks-of 34% in 215 treated patients, comprising 136 patients who received targeted therapies and 79 patients who received immunotherapy. The overall median duration of clinical benefit was 9 months (95% confidence interval of 8-11 months), including 26 patients who were experiencing ongoing clinical benefit at data cut-off. The potential of the Drug Rediscovery protocol is illustrated by the identification of a successful cohort of patients with microsatellite instable tumours who received nivolumab (clinical benefit rate of 63%), and a cohort of patients with colorectal cancer with relatively low mutational load who experienced only limited clinical benefit from immunotherapy. The Drug Rediscovery protocol facilitates the defined use of approved drugs beyond their labels in rare subgroups of cancer, identifies early signals of activity in these subgroups, accelerates the clinical translation of new insights into the use of anticancer drugs outside of their approved label, and creates a publicly available repository of knowledge for future decision-making.


Assuntos
Antineoplásicos/uso terapêutico , Descoberta de Drogas/métodos , Reposicionamento de Medicamentos/tendências , Neoplasias/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Feminino , Humanos , Imunoterapia , Masculino , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Neoplasias/genética , Nivolumabe/uso terapêutico , Medicina de Precisão , Intervalo Livre de Progressão , Projetos de Pesquisa , Adulto Jovem
6.
Leukemia ; 32(4): 931-940, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28972594

RESUMO

RAS pathway mutations have been linked to relapse and chemotherapy resistance in pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL). However, comprehensive data on the frequency and prognostic value of subclonal mutations in well-defined subgroups using highly sensitive and quantitative methods are lacking. Targeted deep sequencing of 13 RAS pathway genes was performed in 461 pediatric BCP-ALL cases at initial diagnosis and in 19 diagnosis-relapse pairs. Mutations were present in 44.2% of patients, with 24.1% carrying a clonal mutation. Mutation frequencies were highest in high hyperdiploid, infant t(4;11)-rearranged, BCR-ABL1-like and B-other cases (50-70%), whereas mutations were less frequent in ETV6-RUNX1-rearranged, and rare in TCF3-PBX1- and BCR-ABL1-rearranged cases (27-4%). RAS pathway-mutated cells were more resistant to prednisolone and vincristine ex vivo. Clonal, but not subclonal, mutations were linked to unfavorable outcome in standard- and high-risk-treated patients. At relapse, most RAS pathway mutations were clonal (9 of 10). RAS mutant cells were sensitive to the MEK inhibitor trametinib ex vivo, and trametinib sensitized resistant cells to prednisolone. We conclude that RAS pathway mutations are frequent, and that clonal, but not subclonal, mutations are associated with unfavorable risk parameters in newly diagnosed pediatric BCP-ALL. These mutations may designate patients eligible for MEK inhibitor treatment.


Assuntos
Linfócitos B/metabolismo , Biomarcadores Tumorais/genética , Mutação/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Proteínas ras/genética , Adolescente , Animais , Linhagem Celular Tumoral , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Camundongos , Camundongos Endogâmicos NOD , Taxa de Mutação , Proteínas de Fusão Oncogênica/genética , Prognóstico , Transdução de Sinais/genética
7.
Ann Oncol ; 28(12): 3070-3075, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-29045504

RESUMO

BACKGROUND: Due to rapid technical advances, steeply declining sequencing costs, and the ever-increasing number of targeted therapies, it can be expected that extensive tumor sequencing such as whole-exome and whole-genome sequencing will soon be applied in standard care. Clinicians will thus be confronted with increasingly complex genetic information and multiple test-platforms to choose from. General medical training, meanwhile, can hardly keep up with the pace of innovation. Consequently, there is a rapidly growing gap between clinical knowledge and genetic potential in cancer care. Multidisciplinary Molecular Tumor Boards (MTBs) have been suggested as a means to address this disparity, but shared experiences are scarce in literature and no quality requirements or guidelines have been published to date. METHODS: Based on literature review, a survey among hospitals in The Netherlands, and our own experience with the establishment of a nationally operating MTB, this article evaluates current knowledge and unmet needs and lays out a strategy for successful MTB implementation. RESULTS: Having access to an MTB can improve and increase the application of genetics-guided cancer care. In our survey, however, <50% of hospitals and only 5% of nonacademic hospitals had access to an MTB. In addition, current MTBs vary widely in terms of composition, tasks, tools, and workflow. This may not only lead to variation in quality of care but also hinders data sharing and thus creation of an effective learning community. CONCLUSIONS: This article acknowledges a leading role for MTBs to govern (extensive) tumor sequencing into daily practice and proposes three basic necessities for successful MTB implementation: (i) global harmonization in cancer sequencing practices and procedures, (ii) minimal member and operational requirements, and (iii) an appropriate unsolicited findings policy. Meeting these prerequisites would not only optimize MTB functioning but also improve general interpretation and application of genomics-guided cancer care.


Assuntos
Genômica/métodos , Oncologia/métodos , Neoplasias/genética , Testes Genéticos , Humanos , Neoplasias/terapia , Países Baixos
8.
Acta Physiol (Oxf) ; 216(3): 358-75, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26495902

RESUMO

AIM: Blocking of lysophosphatidic acid (LPA) receptor (LPAR) 1 may be a novel therapeutic option for bronchopulmonary dysplasia (BPD) by preventing the LPAR1-mediated adverse effects of its ligand (LPA), consisting of lung inflammation, pulmonary arterial hypertension (PAH) and fibrosis. METHODS: In Wistar rats with experimental BPD, induced by continuous exposure to 100% oxygen for 10 days, we determined the beneficial effects of LPAR1 deficiency in neonatal rats with a missense mutation in cytoplasmic helix 8 of LPAR1 and of LPAR1 and -3 blocking with Ki16425. Parameters investigated included survival, lung and heart histopathology, fibrin and collagen deposition, vascular leakage and differential mRNA expression in the lungs of key genes involved in LPA signalling and BPD pathogenesis. RESULTS: LPAR1-mutant rats were protected against experimental BPD and mortality with reduced alveolar septal thickness, lung inflammation (reduced influx of macrophages and neutrophils, and CINC1 expression) and collagen III deposition. However, LPAR1-mutant rats were not protected against alveolar enlargement, increased medial wall thickness of small arterioles, fibrin deposition and vascular alveolar leakage. Treatment of experimental BPD with Ki16425 confirmed the data observed in LPAR1-mutant rats, but did not reduce the pulmonary influx of neutrophils, CINC1 expression and mortality in rats with experimental BPD. In addition, Ki16425 treatment protected against PAH and right ventricular hypertrophy. CONCLUSION: LPAR1 deficiency attenuates pulmonary injury by reducing pulmonary inflammation and fibrosis, thereby reducing mortality, but does not affect alveolar and vascular development and, unlike Ki16425 treatment, does not prevent PAH in neonatal rats with experimental BPD.


Assuntos
Displasia Broncopulmonar/metabolismo , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Receptores de Ácidos Lisofosfatídicos/deficiência , Animais , Animais Recém-Nascidos , Western Blotting , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Hiperóxia/complicações , Isoxazóis/farmacologia , Propionatos/farmacologia , Ratos , Ratos Mutantes , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
9.
Mol Cell Biol ; 21(22): 7587-600, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11604495

RESUMO

Tumor suppressor gene inactivation is a crucial event in oncogenesis. Gene inactivation mechanisms include events resulting in loss of heterozygosity (LOH), gene mutation, and transcriptional silencing. The contribution of each of these different pathways varies among tumor suppressor genes and by cancer type. The factors that influence the relative utilization of gene inactivation pathways are poorly understood. In this study, we describe a detailed quantitative analysis of the three major gene inactivation mechanisms for a model gene at two different genomic integration sites in mouse embryonic stem (ES) cells. In addition, we targeted the major DNA methyltransferase gene, Dnmt1, to investigate the relative contribution of DNA methylation to these various competing gene inactivation pathways. Our data show that gene loss is the predominant mode of inactivation of a herpes simplex virus thymidine kinase neomycin phosphotransferase reporter gene (HSV-TKNeo) at the two integration sites tested and that this event is significantly reduced in Dnmt1-deficient cells. Gene silencing by promoter methylation requires Dnmt1, suggesting that the expression of Dnmt3a and Dnmt3b alone in ES cells is insufficient to achieve effective gene silencing. We used a novel assay to show that missense mutation rates are also substantially reduced in Dnmt1-deficient cells. This is the first direct demonstration that DNA methylation affects point mutation rates in mammalian cells. Surprisingly, the fraction of CpG transition mutations was not reduced in Dnmt1-deficient cells. Finally, we show that methyl group-deficient growth conditions do not cause an increase in missense mutation rates in Dnmt1-proficient cells, as predicted by methyltransferase-mediated mutagenesis models. We conclude that Dnmt1 deficiency and the accompanying genomic DNA hypomethylation result in a reduction of three major pathways of gene inactivation in our model system.


Assuntos
DNA (Citosina-5-)-Metiltransferases/fisiologia , Metilação de DNA , Inativação Gênica , Perda de Heterozigosidade , Mutação de Sentido Incorreto , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Herpesvirus Humano 1/enzimologia , Humanos , Canamicina Quinase/genética , Modelos Genéticos , Células-Tronco/citologia , Timidina Quinase/genética
10.
Eur J Cell Biol ; 79(4): 283-93, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10826496

RESUMO

The small adaptor protein RIL consists of two segments, the C-terminal LIM and the N-terminal PDZ domain, which mediate multiple protein-protein interactions. The RIL LIM domain can interact with PDZ domains in the protein tyrosine phosphatase PTP-BL and with the PDZ domain of RIL itself. Here, we describe and characterise the interaction of the RIL PDZ domain with the zyxin-related protein TRIP6, a protein containing three C-terminal LIM domains. The second LIM domain in TRIP6 is sufficient for a strong interaction with RIL. A weaker interaction with the third LIM domain in TRIP6, including the proper C-terminus, is also evident. TRIP6 also interacts with the second out of five PDZ motifs in PTP-BL. For this interaction to occur both the third LIM domain and the proper C-terminus are necessary. RNA expression analysis revealed overlapping patterns of expression for TRIP6, RIL and PTP-BL, most notably in tissues of epithelial origin. Furthermore, in transfected epithelial cells TRIP6 can be co-precipitated with RIL and PTP-BL PDZ polypeptides, and a co-localisation of TRIP6 and RIL with Factin structures is evident. Taken together, PTP-BL, RIL and TRIP6 may function as components of multi-protein complexes at actin-based sub-cellular structures.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Ligação a DNA/metabolismo , Metaloproteínas/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Fatores de Transcrição/metabolismo , ATPases Associadas a Diversas Atividades Celulares , Actinas/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Western Blotting , Células CACO-2 , Clonagem Molecular , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/química , Imunofluorescência , Biblioteca Gênica , Glicoproteínas , Humanos , Hibridização In Situ , Proteínas com Domínio LIM , Camundongos , Proteínas dos Microfilamentos , Dados de Sequência Molecular , Plasmídeos , Testes de Precipitina , Complexo de Endopeptidases do Proteassoma , Ligação Proteica , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 13 , Proteínas Tirosina Fosfatases/química , RNA Mensageiro/metabolismo , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Fatores de Transcrição/química , Transfecção , Técnicas do Sistema de Duplo-Híbrido , Zixina
11.
J Cell Sci ; 112 ( Pt 19): 3299-308, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10504335

RESUMO

PTP-BL is a cytosolic multidomain protein tyrosine phosphatase that shares homologies with several submembranous and tumor suppressor proteins. Here we show, by transient expression of modular protein domains of PTP-BL in epithelial MDCK cells, that the presence of a FERM domain in the protein is both necessary and sufficient for its targeting to the apical side of epithelial cells. Furthermore, immuno-electron microscopy on stable expressing MDCK pools, that were obtained using an EGFP-based cell sorting protocol, revealed that FERM domain containing fusion proteins are enriched in microvilli and have a typical submembranous location at about 10-15 nm from the plasma membrane. Immunofluorescence microscopy suggested colocalization of the FERM domain moiety with the membrane-cytoskeleton linker ezrin. However, at the electron microscopy level this colocalization cannot be confirmed nor can we detect a direct interaction by immunoprecipitation assays. Fluorescence recovery after photobleaching (FRAP) experiments show that PTP-BL confinement is based on a dynamic steady state and that complete redistribution of the protein may occur within 20 minutes. Our observations suggest that relocation is mediated via a cytosolic pool, rather than by lateral movement. Finally, we show that PTP-BL phosphatase domains are involved in homotypic interactions, as demonstrated by yeast two-hybrid assays. Both the highly restricted subcellular compartmentalization and its specific associative properties may provide the appropriate conditions for regulating substrate specificity and catalytic activity of this member of the PTP family.


Assuntos
Polaridade Celular/fisiologia , Proteínas do Citoesqueleto , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Proteínas de Neoplasias/química , Neuropeptídeos , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/metabolismo , Animais , Transporte Biológico/fisiologia , Células COS , Domínio Catalítico/fisiologia , Compartimento Celular/fisiologia , Dimerização , Ativação Enzimática/fisiologia , Células Epiteliais/ultraestrutura , Rim/citologia , Proteínas de Membrana/química , Microscopia de Fluorescência , Microscopia Imunoeletrônica , Mutagênese/fisiologia , Proteínas de Neoplasias/genética , Fosfoproteínas/metabolismo , Plasmídeos , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 13 , Proteínas Tirosina Fosfatases/genética , Técnicas do Sistema de Duplo-Híbrido
12.
FEBS Lett ; 459(3): 291-8, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10526152

RESUMO

We here describe the identification and characterization of a novel bromodomain-containing protein, the bromodomain protein of 75 kDa (BP75). Initially, we identified BP75 in a two-hybrid screening for proteins that interact with the first PDZ (acronym for post-synaptic density protein PSD-95, Drosophila discs large tumor suppressor DlgA and the tight junction protein ZO-1) domain in protein tyrosine phosphatase-BAS-like (PTP-BL). We found that BP75 is expressed ubiquitously and show that both BP75 and a PTP-BL deletion mutant consisting of the first PDZ domain are located mainly in the nucleus, although cytoplasmic localization is also evident. Full-length PTP-BL, on the contrary, is predominantly localized in the cytoplasm, although some basal nuclear staining is observed. The described molecular interaction may reflect a mechanism of coupling submembraneous signalling events and nuclear events.


Assuntos
Bromo/química , Proteínas de Transporte/genética , Proteínas Cromossômicas não Histona , Sequência Conservada , Proteínas Nucleares/genética , Células 3T3 , Sequência de Aminoácidos , Animais , Células COS , Proteínas de Transporte/isolamento & purificação , Proteínas de Transporte/metabolismo , DNA Complementar/metabolismo , Proteína 4 Homóloga a Disks-Large , Guanilato Quinases , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/isolamento & purificação , Proteínas Nucleares/metabolismo , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 13 , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/metabolismo , Homologia de Sequência de Aminoácidos , Frações Subcelulares/metabolismo , Técnicas do Sistema de Duplo-Híbrido
13.
J Biol Chem ; 272(48): 30215-20, 1997 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-9374505

RESUMO

Recently, one of the PDZ domains in the cytosolic protein-tyrosine phosphatase Fas-associated phosphatase-1 (FAP-1)/protein-tyrosine phosphatase-BAS (PTP-BAS) was shown to interact with the carboxyl-terminal tS-L-V peptide of the human Fas receptor (Sato, T., Irie, S., Kitada, S., and Reed, J. C. (1995) Science 268, 411-415), suggesting a role for protein (de)phosphorylation in Fas signaling. To investigate whether this interaction is conserved in mouse, we performed yeast two-hybrid interaction experiments and transfection studies in mouse T cell lines. For the corresponding PDZ motif in the mouse homologue of FAP-1/PTP-BAS, protein-tyrosine phosphatase-BAS-like (PTP-BL), only an interaction with human but not with mouse Fas could be detected. Presence of the tS-L-V motif proper, which is unique for human Fas, rather than the structural context of its carboxyl terminus, apparently explains the initially observed binding. To test for functional conservation of any indirect involvement of PTP-BL in Fas-mediated signaling, we generated T lymphoma cell lines stably expressing mouse or human Fas receptor with and without PTP-BL. No inhibitory effect of PTP-BL was observed upon triggering apoptosis using mouse or human Fas-activating antibodies. Together with the markedly different tissue expression patterns for PTP-BL and Fas receptor, our findings suggest that protein-tyrosine phosphatase PTP-BL does not play a key role in the Fas-mediated death pathway.


Assuntos
Apoptose , Proteínas de Transporte/metabolismo , Glicoproteínas de Membrana/fisiologia , Proteínas Tirosina Fosfatases/metabolismo , Receptor fas/fisiologia , Animais , Sítios de Ligação , Bovinos , Linhagem Celular , Proteína Ligante Fas , Expressão Gênica , Humanos , Camundongos , Ligação Proteica , Proteína Fosfatase 1 , Proteína Tirosina Fosfatase não Receptora Tipo 13 , RNA Mensageiro/genética , Ratos , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Distribuição Tecidual
14.
FEBS Lett ; 409(1): 53-6, 1997 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-9199503

RESUMO

PDZ motifs are small protein-protein interaction modules that are thought to play a role in the clustering of submembranous signalling molecules. The specificity and functional consequences of their associative actions is still largely unknown. Using two-hybrid methodology we here demonstrate that the PDZ motif of neuronal nitric oxide synthase (nNOS) can mediate the binding to several other proteins in brain. Peptide library screening showed that proteins bearing a carboxy-terminal G(D,E)XV* sequence are preferred targets for the nNOS amino-terminal PDZ motif. Potential nNOS targets include a melanoma-associated antigen, cyclophilins and the alpha1C-adrenergic receptor.


Assuntos
Sequência de Aminoácidos , Neurônios/enzimologia , Óxido Nítrico Sintase/química , Óxido Nítrico Sintase/metabolismo , Animais , Encéfalo/enzimologia , Encéfalo/metabolismo , Clonagem Molecular , Sequência Consenso/genética , Humanos , Camundongos , Neurônios/química , Neurônios/metabolismo , Óxido Nítrico Sintase/genética , Biblioteca de Peptídeos , Ligação Proteica , Proteínas Recombinantes/síntese química , Proteínas Recombinantes/metabolismo , Frações Subcelulares/química , Frações Subcelulares/enzimologia , Frações Subcelulares/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA