Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Mol Cell ; 81(21): 4440-4456.e7, 2021 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-34597596

RESUMO

Protection of stalled replication forks is critical to genomic stability. Using genetic and proteomic analyses, we discovered the Protexin complex containing the ssDNA binding protein SCAI and the DNA polymerase REV3. Protexin is required specifically for protecting forks stalled by nucleotide depletion, fork barriers, fragile sites, and DNA inter-strand crosslinks (ICLs), where it promotes homologous recombination and repair. Protexin loss leads to ssDNA accumulation and profound genomic instability in response to ICLs. Protexin interacts with RNA POL2, and both oppose EXO1's resection of DNA on forks remodeled by the FANCM translocase activity. This pathway acts independently of BRCA/RAD51-mediated fork stabilization, and cells with BRCA2 mutations were dependent on SCAI for survival. These data suggest that Protexin and its associated factors establish a new fork protection pathway that counteracts fork resection in part through a REV3 polymerase-dependent resynthesis mechanism of excised DNA, particularly at ICL stalled forks.


Assuntos
Proteína BRCA2/metabolismo , DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/química , DNA Polimerase Dirigida por DNA/química , Exodesoxirribonucleases/metabolismo , Fatores de Transcrição/química , Animais , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Reparo do DNA , DNA de Cadeia Simples/química , DNA de Cadeia Simples/metabolismo , Células HeLa , Humanos , Ácido Mevalônico , Camundongos , Complexos Multiproteicos , Mutação , Ligação Proteica , Conformação Proteica , RNA Guia de Cinetoplastídeos/metabolismo , RNA Interferente Pequeno/metabolismo , Recombinação Genética
2.
Cell ; 147(2): 459-74, 2011 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-21963094

RESUMO

Cullin-RING ligases (CRLs) represent the largest E3 ubiquitin ligase family in eukaryotes, and the identification of their substrates is critical to understanding regulation of the proteome. Using genetic and pharmacologic Cullin inactivation coupled with genetic (GPS) and proteomic (QUAINT) assays, we have identified hundreds of proteins whose stabilities or ubiquitylation status are regulated by CRLs. Together, these approaches yielded many known CRL substrates as well as a multitude of previously unknown putative substrates. We demonstrate that one substrate, NUSAP1, is an SCF(Cyclin F) substrate during S and G2 phases of the cell cycle and is also degraded in response to DNA damage. This collection of regulated substrates is highly enriched for nodes in protein interaction networks, representing critical connections between regulatory pathways. This demonstrates the broad role of CRL ubiquitylation in all aspects of cellular biology and provides a set of proteins likely to be key indicators of cellular physiology.


Assuntos
Genoma Humano , Proteoma/análise , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Ciclopentanos/farmacologia , Inibidores Enzimáticos/farmacologia , Humanos , Pirimidinas/farmacologia , Ubiquitina-Proteína Ligases/genética
3.
Mol Cell ; 60(2): 280-93, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26474068

RESUMO

We have used quantitative proteomics to profile ubiquitination in the DNA damage response (DDR). We demonstrate that RPA, which functions as a protein scaffold in the replication stress response, is multiply ubiquitinated upon replication fork stalling. Ubiquitination of RPA occurs on chromatin, involves sites outside its DNA binding channel, does not cause proteasomal degradation, and increases under conditions of fork collapse, suggesting a role in repair at stalled forks. We demonstrate that the E3 ligase RFWD3 mediates RPA ubiquitination. RFWD3 is necessary for replication fork restart, normal repair kinetics during replication stress, and homologous recombination (HR) at stalled replication forks. Mutational analysis suggests that multisite ubiquitination of the entire RPA complex is responsible for repair at stalled forks. Multisite protein group sumoylation is known to promote HR in yeast. Our findings reveal a similar requirement for multisite protein group ubiquitination during HR at stalled forks in mammalian cells.


Assuntos
Reparo do DNA , Replicação do DNA , DNA/genética , Subunidades Proteicas/genética , Proteína de Replicação A/genética , Ubiquitina-Proteína Ligases/genética , Cromatina/química , Cromatina/metabolismo , DNA/química , Dano ao DNA , Células HeLa , Recombinação Homóloga , Humanos , Modelos Moleculares , Mutação , Ligação Proteica , Subunidades Proteicas/metabolismo , Proteína de Replicação A/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
4.
Mol Cell ; 59(5): 867-81, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26051181

RESUMO

Execution of the DNA damage response (DDR) relies upon a dynamic array of protein modifications. Using quantitative proteomics, we have globally profiled ubiquitination, acetylation, and phosphorylation in response to UV and ionizing radiation. To improve acetylation site profiling, we developed the strategy FACET-IP. Our datasets of 33,500 ubiquitination and 16,740 acetylation sites provide valuable insight into DDR remodeling of the proteome. We find that K6- and K33-linked polyubiquitination undergo bulk increases in response to DNA damage, raising the possibility that these linkages are largely dedicated to DDR function. We also show that Cullin-RING ligases mediate 10% of DNA damage-induced ubiquitination events and that EXO1 is an SCF-Cyclin F substrate in the response to UV radiation. Our extensive datasets uncover additional regulated sites on known DDR players such as PCNA and identify previously unknown DDR targets such as CENPs, underscoring the broad impact of the DDR on cellular physiology.


Assuntos
Dano ao DNA , Proteômica/métodos , Acetilação/efeitos da radiação , Proteínas Culina/metabolismo , Reparo do DNA , Enzimas Reparadoras do DNA/metabolismo , Bases de Dados de Proteínas , Exodesoxirribonucleases/metabolismo , Células HeLa , Humanos , Fosforilação/efeitos da radiação , Complexo de Endopeptidases do Proteassoma/metabolismo , Análise Serial de Proteínas/estatística & dados numéricos , Proteoma/metabolismo , Proteoma/efeitos da radiação , Proteômica/estatística & dados numéricos , Fuso Acromático/metabolismo , Ubiquitinação/efeitos da radiação
5.
Proc Natl Acad Sci U S A ; 113(26): E3676-85, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27298342

RESUMO

The cellular response to DNA damage during S-phase regulates a complicated network of processes, including cell-cycle progression, gene expression, DNA replication kinetics, and DNA repair. In fission yeast, this S-phase DNA damage response (DDR) is coordinated by two protein kinases: Rad3, the ortholog of mammalian ATR, and Cds1, the ortholog of mammalian Chk2. Although several critical downstream targets of Rad3 and Cds1 have been identified, most of their presumed targets are unknown, including the targets responsible for regulating replication kinetics and coordinating replication and repair. To characterize targets of the S-phase DDR, we identified proteins phosphorylated in response to methyl methanesulfonate (MMS)-induced S-phase DNA damage in wild-type, rad3∆, and cds1∆ cells by proteome-wide mass spectrometry. We found a broad range of S-phase-specific DDR targets involved in gene expression, stress response, regulation of mitosis and cytokinesis, and DNA replication and repair. These targets are highly enriched for proteins required for viability in response to MMS, indicating their biological significance. Furthermore, the regulation of these proteins is similar in fission and budding yeast, across 300 My of evolution, demonstrating a deep conservation of S-phase DDR targets and suggesting that these targets may be critical for maintaining genome stability in response to S-phase DNA damage across eukaryotes.


Assuntos
Dano ao DNA , Fase S , Schizosaccharomyces/genética , Quinase do Ponto de Checagem 2/genética , Quinase do Ponto de Checagem 2/metabolismo , Dano ao DNA/efeitos dos fármacos , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Instabilidade Genômica/efeitos dos fármacos , Metanossulfonato de Metila/toxicidade , Fase S/efeitos dos fármacos , Schizosaccharomyces/citologia , Schizosaccharomyces/efeitos dos fármacos , Schizosaccharomyces/enzimologia , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo
6.
Proc Natl Acad Sci U S A ; 113(26): E3667-75, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27298372

RESUMO

The DNA damage response (DDR) is regulated by a protein kinase signaling cascade that orchestrates DNA repair and other processes. Identifying the substrate effectors of these kinases is critical for understanding the underlying physiology and mechanism of the response. We have used quantitative mass spectrometry to profile DDR-dependent phosphorylation in budding yeast and genetically explored the dependency of these phosphorylation events on the DDR kinases MEC1, RAD53, CHK1, and DUN1. Based on these screens, a database containing many novel DDR-regulated phosphorylation events has been established. Phosphorylation of many of these proteins has been validated by quantitative peptide phospho-immunoprecipitation and examined for functional relevance to the DDR through large-scale analysis of sensitivity to DNA damage in yeast deletion strains. We reveal a link between DDR signaling and the metabolic pathways of inositol phosphate and phosphatidyl inositol synthesis, which are required for resistance to DNA damage. We also uncover links between the DDR and TOR signaling as well as translation regulation. Taken together, these data shed new light on the organization of DDR signaling in budding yeast.


Assuntos
Dano ao DNA , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Transdução de Sinais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Quinase do Ponto de Checagem 2/genética , Quinase do Ponto de Checagem 2/metabolismo , Reparo do DNA , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
7.
Semin Neurol ; 38(1): 50-61, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29548052

RESUMO

Glioblastoma (GBM) is the most common primary malignant tumor of the central nervous system. The current standard of care for GBM is maximal resection followed by postoperative radiation with concomitant and adjuvant temozolomide. Despite this multimodality treatment, the median survival for GBM remains marginally better than 1 year. In the past decade, genome-wide analyses have uncovered new molecular features of GBM that have refined its classification and provided new insights into the molecular basis for GBM pathogenesis. Here, we review these molecular features and discuss major clinical trials that have recently defined the field. We describe genetic alterations in isocitrate dehydrogenase, ATRX, the telomerase promoter, and histone H3 variants that promote GBM tumorigenesis and have altered GBM categorization. We also discuss intratumoral genetic heterogeneity as one explanation for therapeutic failures and explain how ultra-long extensions of glioma cells, called tumor microtubes, mediate therapeutic resistance. These findings provide new insights into GBM biology and offer hope for the development of next-generation therapies.


Assuntos
Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos
8.
J Virol ; 87(14): 7837-52, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23658454

RESUMO

The interferon-induced transmembrane protein 3 (IFITM3) gene is an interferon-stimulated gene that inhibits the replication of multiple pathogenic viruses in vitro and in vivo. IFITM3 is a member of a large protein superfamily, whose members share a functionally undefined area of high amino acid conservation, the CD225 domain. We performed mutational analyses of IFITM3 and identified multiple residues within the CD225 domain, consisting of the first intramembrane domain (intramembrane domain 1 [IM1]) and a conserved intracellular loop (CIL), that are required for restriction of both influenza A virus (IAV) and dengue virus (DENV) infection in vitro. Two phenylalanines within IM1 (F75 and F78) also mediate a physical association between IFITM proteins, and the loss of this interaction decreases IFITM3-mediated restriction. By extension, similar IM1-mediated associations may contribute to the functions of additional members of the CD225 domain family. IFITM3's distal N-terminal domain is also needed for full antiviral activity, including a tyrosine (Y20), whose alteration results in mislocalization of a portion of IFITM3 to the cell periphery and surface. Comparative analyses demonstrate that similar molecular determinants are needed for IFITM3's restriction of both IAV and DENV. However, a portion of the CIL including Y99 and R87 is preferentially needed for inhibition of the orthomyxovirus. Several IFITM3 proteins engineered with rare single-nucleotide polymorphisms demonstrated reduced expression or mislocalization, and these events were associated with enhanced viral replication in vitro, suggesting that possessing such alleles may impact an individual's risk for viral infection. On the basis of this and other data, we propose a model for IFITM3-mediated restriction.


Assuntos
Vírus da Dengue/fisiologia , Vírus da Influenza A/fisiologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Replicação Viral/fisiologia , Sequência de Aminoácidos , Animais , Técnicas de Cultura de Células , Clonagem Molecular , Sequência Conservada/genética , Análise Mutacional de DNA , DNA Complementar/genética , Cães , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Imunoprecipitação , Células Madin Darby de Rim Canino , Espectrometria de Massas , Microscopia Confocal , Modelos Biológicos , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único/genética , Estrutura Terciária de Proteína/genética , Replicação Viral/genética
9.
Proc Natl Acad Sci U S A ; 108(24): 9845-50, 2011 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-21628590

RESUMO

The anaphase-promoting complex/cyclosome (APC/C) is a cell cycle-regulated E3 ubiquitin ligase that controls the degradation of substrate proteins at mitotic exit and throughout the G1 phase. We have identified an APC/C substrate and cell cycle-regulated protein, KIAA0101/PAF15. PAF15 protein levels peak in the G2/M phase of the cell cycle and drop rapidly at mitotic exit in an APC/C- and KEN-box-dependent fashion. PAF15 associates with proliferating cell nuclear antigen (PCNA), and depletion of PAF15 decreases the number of cells in S phase, suggesting a role for it in cell cycle regulation. Following irradiation, PAF15 colocalized with γH2AX foci at sites of DNA damage through its interaction with PCNA. Finally, PAF15 depletion led to an increase in homologous recombination-mediated DNA repair, and overexpression caused sensitivity to UV-induced DNA damage. We conclude that PAF15 is an APC/C-regulated protein involved in both cell cycle progression and the DNA damage response.


Assuntos
Proteínas de Transporte/metabolismo , Ciclo Celular , Antígeno Nuclear de Célula em Proliferação/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Western Blotting , Proteínas de Transporte/genética , Divisão Celular , Linhagem Celular , Linhagem Celular Tumoral , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA , Citometria de Fluxo , Fase G2 , Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Ligação Proteica , Interferência de RNA , Recombinação Genética , Fase S , Especificidade por Substrato , Ubiquitinação
10.
Nat Genet ; 33(2): 129-37, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12514735

RESUMO

Several proteins implicated in the pathogenesis of polycystic kidney disease (PKD) localize to cilia. Furthermore, cilia are malformed in mice with PKD with mutations in TgN737Rpw (encoding polaris). It is not known, however, whether ciliary dysfunction occurs or is relevant to cyst formation in PKD. Here, we show that polycystin-1 (PC1) and polycystin-2 (PC2), proteins respectively encoded by Pkd1 and Pkd2, mouse orthologs of genes mutated in human autosomal dominant PKD, co-distribute in the primary cilia of kidney epithelium. Cells isolated from transgenic mice that lack functional PC1 formed cilia but did not increase Ca(2+) influx in response to physiological fluid flow. Blocking antibodies directed against PC2 similarly abolished the flow response in wild-type cells as did inhibitors of the ryanodine receptor, whereas inhibitors of G-proteins, phospholipase C and InsP(3) receptors had no effect. These data suggest that PC1 and PC2 contribute to fluid-flow sensation by the primary cilium in renal epithelium and that they both function in the same mechanotransduction pathway. Loss or dysfunction of PC1 or PC2 may therefore lead to PKD owing to the inability of cells to sense mechanical cues that normally regulate tissue morphogenesis.


Assuntos
Cálcio/metabolismo , Cílios/fisiologia , Epitélio/metabolismo , Homeostase/fisiologia , Proteínas de Membrana/fisiologia , Rim Policístico Autossômico Dominante/fisiopatologia , Proteínas/fisiologia , Animais , Cafeína/farmacologia , Canais de Cálcio/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Heterozigoto , Humanos , Rim/metabolismo , Camundongos , Camundongos Knockout , Mutação , Ligação Proteica , Transporte Proteico , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Transdução de Sinais/fisiologia , Canais de Cátion TRPP , Tubulina (Proteína)/metabolismo
11.
J Cell Biol ; 222(5)2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37036693

RESUMO

Replication fork reversal is an important mechanism to protect the stability of stalled forks and thereby preserve genomic integrity. While multiple enzymes have been identified that can remodel forks, their regulation remains poorly understood. Here, we demonstrate that the ubiquitin ligase RFWD3, whose mutation causes Fanconi Anemia, promotes recruitment of the DNA translocase ZRANB3 to stalled replication forks and ubiquitinated sites of DNA damage. Using electron microscopy, we show that RFWD3 stimulates fork remodeling in a ZRANB3-epistatic manner. Fork reversal is known to promote nascent DNA degradation in BRCA2-deficient cells. Consistent with a role for RFWD3 in fork reversal, inactivation of RFWD3 in these cells rescues fork degradation and collapse, analogous to ZRANB3 inactivation. RFWD3 loss impairs ZRANB3 localization to spontaneous nuclear foci induced by inhibition of the PCNA deubiquitinase USP1. We demonstrate that RFWD3 promotes PCNA ubiquitination and interaction with ZRANB3, providing a mechanism for RFWD3-dependent recruitment of ZRANB3. Together, these results uncover a new role for RFWD3 in regulating ZRANB3-dependent fork remodeling.


Assuntos
DNA Helicases , Replicação do DNA , DNA , Ubiquitina-Proteína Ligases , DNA/genética , Dano ao DNA , Replicação do DNA/genética , Proteínas de Ligação a DNA/metabolismo , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Humanos , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Ubiquitinação
12.
Breast Cancer Res ; 14(2): 306, 2012 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-22494569

RESUMO

BRCA1 is a tumor suppressor with critical roles in the maintenance of genomic stability. It encodes a large protein with an amino-terminal RING domain that possesses ubiquitin-ligase activity. Given the occurrence of numerous cancer-causing mutations within its RING domain, investigators have long suspected that BRCA1's ubiquitin ligase is important for its tumor suppression and DNA repair activities. Using genetically engineered mouse models, two recent studies shed light on this age-old hypothesis.


Assuntos
Proteína BRCA1/química , Proteína BRCA1/genética , Genes Supressores de Tumor , Animais , Proteína BRCA1/metabolismo , Neoplasias da Mama/genética , Reparo do DNA , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Estrutura Terciária de Proteína
13.
J Cell Biol ; 219(6)2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32391871

RESUMO

BRCA1/2 help maintain genomic integrity by stabilizing stalled forks. Here, we identify the E3 ligase RFWD3 as an essential modulator of stalled fork stability in BRCA2-deficient cells and show that codepletion of RFWD3 rescues fork degradation, collapse, and cell sensitivity upon replication stress. Stalled forks in BRCA2-deficient cells accumulate phosphorylated and ubiquitinated replication protein A (ubq-pRPA), the latter of which is mediated by RFWD3. Generation of this intermediate requires SMARCAL1, suggesting that it depends on stalled fork reversal. We show that in BRCA2-deficient cells, rescuing fork degradation might not be sufficient to ensure fork repair. Depleting MRE11 in BRCA2-deficient cells does block fork degradation, but it does not prevent fork collapse and cell sensitivity in the presence of replication stress. No such ubq-pRPA intermediate is formed in BRCA1-deficient cells, and our results suggest that BRCA1 may function upstream of BRCA2 in the stalled fork repair pathway. Collectively, our data uncover a novel mechanism by which RFWD3 destabilizes forks in BRCA2-deficient cells.


Assuntos
Proteína BRCA2/metabolismo , Dano ao DNA/genética , Reparo do DNA/genética , Replicação do DNA/genética , Proteína de Replicação A/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteína BRCA1/deficiência , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/deficiência , Proteína BRCA2/genética , Ciclo Celular , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Dano ao DNA/efeitos dos fármacos , DNA Helicases/genética , DNA Helicases/metabolismo , Replicação do DNA/efeitos dos fármacos , Humanos , Hidroxiureia/farmacologia , Proteína Homóloga a MRE11/deficiência , Proteína Homóloga a MRE11/genética , Proteína Homóloga a MRE11/metabolismo , Mutação , Fosforilação , RNA Interferente Pequeno , Rad51 Recombinase/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitinação/genética
14.
Neuro Oncol ; 20(2): 174-183, 2018 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-29016815

RESUMO

Management of glioblastoma in the elderly population is challenging. In the near future, more than half of patients with this tumor will be over the age of 65. Clinicians have been historically reluctant to treat such patients with the same intensity as younger patients. Due to upper age limits or poor accrual of elderly patients in clinical trials, randomized data for this patient population have been relatively sparse until recently. In this review, we will discuss the concept of an elderly patient population, describe evidence for molecular differences in glioblastoma of elderly versus young patients, evaluate recent first-line trials studying glioblastoma in elderly patients, and discuss best therapeutic practices including the value of molecular testing.


Assuntos
Envelhecimento/fisiologia , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Temozolomida/uso terapêutico , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/patologia , Glioblastoma/diagnóstico , Glioblastoma/patologia , Humanos , Resultado do Tratamento
15.
Neurosurg Focus ; 23(4): E5, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17961042

RESUMO

Meningiomas are the second most common primary tumor of the brain. Gross-total resection remains the preferred treatment if achievable with minimal morbidity. For incompletely resected or inoperable benign meningiomas, 3D conformal external-beam radiation therapy can provide durable local tumor control in 90 to 95% of cases. Stereotactic radiosurgery (SRS) and fractionated stereotactic radiotherapy (SRT) are highly conformal techniques, using steep dose gradients and stereotactic patient immobilization. Stereotactic radiosurgery has been used as an alternative or adjuvant therapy to surgery for meningiomas in locations, such as the skull base, where operative manipulation may be particularly difficult. Stereotactic radiotherapy is useful for larger meningiomas (> 3-3.5 cm) and those closely approximating critical structures, such as the optic chiasm and brainstem. Although SRS has longer follow-up than SRT, both techniques have excellent 5-year tumor control rates of greater than 90% for benign meningiomas. Stereotactic radiotherapy has toxicity equivalent to that of radiosurgery, despite its biased use for larger meningiomas with more complicated volumes. Reported rates of imaging-documented regression are higher for radiosurgery, but neurological recovery is relatively good with both techniques. Stereotactic radiosurgery and fractionated SRT are complementary techniques appropriate for different clinical scenarios.


Assuntos
Neoplasias Meníngeas/patologia , Neoplasias Meníngeas/cirurgia , Meningioma/patologia , Meningioma/cirurgia , Radiocirurgia , Fracionamento da Dose de Radiação , Humanos , Neoplasias Meníngeas/radioterapia , Meningioma/radioterapia
16.
BMC Biochem ; 6: 22, 2005 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-16242037

RESUMO

BACKGROUND: The mitotic exit network (MEN) is a group of proteins that form a signaling cascade that is essential for cells to exit mitosis in Saccharomyces cerevisiae. The MEN has also been implicated in playing a role in cytokinesis. Two components of this signaling pathway are the protein kinase Dbf2 and its binding partner essential for its kinase activity, Mob1. The components of MEN that act upstream of Dbf2-Mob1 have been characterized, but physiological substrates for Dbf2-Mob1 have yet to be identified. RESULTS: Using a combination of peptide library selection, phosphorylation of optimal peptide variants, and screening of a phosphosite array, we found that Dbf2-Mob1 preferentially phosphorylated serine over threonine and required an arginine three residues upstream of the phosphorylated serine in its substrate. This requirement for arginine in peptide substrates could not be substituted with the similarly charged lysine. This specificity determined for peptide substrates was also evident in many of the proteins phosphorylated by Dbf2-Mob1 in a proteome chip analysis. CONCLUSION: We have determined by peptide library selection and phosphosite array screening that the protein kinase Dbf2-Mob1 preferentially phosphorylated substrates that contain an RXXS motif. A subsequent proteome microarray screen revealed proteins that can be phosphorylated by Dbf2-Mob1 in vitro. These proteins are enriched for RXXS motifs, and may include substrates that mediate the function of Dbf2-Mob1 in mitotic exit and cytokinesis. The relatively low degree of sequence restriction at the site of phosphorylation suggests that Dbf2 achieves specificity by docking its substrates at a site that is distinct from the phosphorylation site.


Assuntos
Biblioteca de Peptídeos , Análise Serial de Proteínas/métodos , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteoma/genética , Proteoma/metabolismo , Sequência de Aminoácidos , Dados de Sequência Molecular , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidade por Substrato/fisiologia
17.
Elife ; 2: e00358, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23682311

RESUMO

During all stages of tumor progression, cancer cells are subjected to inappropriate extracellular matrix environments and must undergo adaptive changes in order to evade growth constraints associated with the loss of matrix attachment. A gain of function screen for genes that enable proliferation independently of matrix anchorage identified a cell adhesion molecule PVRL4 (poliovirus-receptor-like 4), also known as Nectin-4. PVRL4 promotes anchorage-independence by driving cell-to-cell attachment and matrix-independent integrin ß4/SHP-2/c-Src activation. Solid tumors frequently have copy number gains of the PVRL4 locus and some have focal amplifications. We demonstrate that the transformation of breast cancer cells is dependent on PVRL4. Furthermore, growth of orthotopically implanted tumors in vivo is inhibited by blocking PVRL4-driven cell-to-cell attachment with monoclonal antibodies, demonstrating a novel strategy for targeted therapy of cancer. DOI:http://dx.doi.org/10.7554/eLife.00358.001.


Assuntos
Neoplasias da Mama/metabolismo , Moléculas de Adesão Celular/metabolismo , Comunicação Celular , Transformação Celular Neoplásica/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Adesão Celular , Moléculas de Adesão Celular/antagonistas & inibidores , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/imunologia , Comunicação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Variações do Número de Cópias de DNA , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Integrina beta4/metabolismo , Camundongos Nus , Nectinas , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases da Família src/metabolismo
18.
Nat Struct Mol Biol ; 20(2): 202-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23295261

RESUMO

X-chromosome dosage compensation by the MSL (male-specific lethal) complex is required in Drosophila melanogaster to increase gene expression from the single male X to equal that of both female X chromosomes. Instead of focusing solely on protein complexes released from DNA, here we used chromatin-interacting protein MS (ChIP-MS) to identify MSL interactions on cross-linked chromatin. We identified MSL-enriched histone modifications, including histone H4 Lys16 acetylation and histone H3 Lys36 methylation, and CG4747, a putative Lys36-trimethylated histone H3 (H3K36me3)-binding protein. CG4747 is associated with the bodies of active genes, coincident with H3K36me3, and is mislocalized in the Set2 mutant lacking H3K36me3. CG4747 loss of function in vivo results in partial mislocalization of the MSL complex to autosomes, and RNA interference experiments confirm that CG4747 and Set2 function together to facilitate targeting of the MSL complex to active genes, validating the ChIP-MS approach.


Assuntos
Mecanismo Genético de Compensação de Dose/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Histonas/metabolismo , Espectrometria de Massas/métodos , Oxirredutases/metabolismo , Acetilação , Animais , Animais Geneticamente Modificados , Western Blotting , Imunoprecipitação da Cromatina , Feminino , Histona-Lisina N-Metiltransferase/metabolismo , Masculino , Metilação , Proteínas Nucleares , Interferência de RNA
20.
Mol Cell ; 17(1): 37-48, 2005 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-15629715

RESUMO

The cellular response to DNA damage is mediated by evolutionarily conserved Ser/Thr kinases, phosphorylation of Cdc25 protein phosphatases, binding to 14-3-3 proteins, and exit from the cell cycle. To investigate DNA damage responses mediated by the p38/stress-activated protein kinase (SAPK) axis of signaling, the optimal phosphorylation motifs of mammalian p38alpha SAPK and MAPKAP kinase-2 were determined. The optimal substrate motif for MAPKAP kinase-2, but not for p38 SAPK, closely matches the 14-3-3 binding site on Cdc25B/C. We show that MAPKAP kinase-2 is directly responsible for Cdc25B/C phosphorylation and 14-3-3 binding in vitro and in response to UV-induced DNA damage within mammalian cells. Downregulation of MAPKAP kinase-2 eliminates DNA damage-induced G2/M, G1, and intra S phase checkpoints. We propose that MAPKAP kinase-2 is a new member of the DNA damage checkpoint kinase family that functions in parallel with Chk1 and Chk2 to integrate DNA damage signaling responses and cell cycle arrest in mammalian cells.


Assuntos
Ciclo Celular/fisiologia , Ciclo Celular/efeitos da radiação , Proteínas Quinases/metabolismo , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Apoptose , Sequência de Bases , Domínio Catalítico , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Dano ao DNA , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular , Modelos Biológicos , Modelos Moleculares , Fosforilação , Proteínas Quinases/química , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases , RNA Interferente Pequeno/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Eletricidade Estática , Raios Ultravioleta , Fosfatases cdc25/genética , Fosfatases cdc25/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/química , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa