Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
EBioMedicine ; 40: 77-91, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30594554

RESUMO

BACKGROUND: Meningioma is the most frequent primary intracranial tumour. Surgical resection remains the main therapeutic option as pharmacological intervention is hampered by poor knowledge of their proteomic signature. There is an urgent need to identify new therapeutic targets and biomarkers of meningioma. METHODS: We performed proteomic profiling of grade I, II and III frozen meningioma specimens and three normal healthy human meninges using LC-MS/MS to analyse global proteins, enriched phosphoproteins and phosphopeptides. Differential expression and functional annotation of proteins was completed using Perseus, IPA® and DAVID. We validated differential expression of proteins and phosphoproteins by Western blot on a meningioma validation set and by immunohistochemistry. FINDINGS: We quantified 3888 proteins and 3074 phosphoproteins across all meningioma grades and normal meninges. Bioinformatics analysis revealed commonly upregulated proteins and phosphoproteins to be enriched in Gene Ontology terms associated with RNA metabolism. Validation studies confirmed significant overexpression of proteins such as EGFR and CKAP4 across all grades, as well as the aberrant activation of the downstream PI3K/AKT pathway, which seems differential between grades. Further, we validated upregulation of the total and activated phosphorylated form of the NIMA-related kinase, NEK9, involved in mitotic progression. Novel proteins identified and validated in meningioma included the nuclear proto-oncogene SET, the splicing factor SF2/ASF and the higher-grade specific protein, HK2, involved in cellular metabolism. INTERPRETATION: Overall, we generated a proteomic thesaurus of meningiomas for the identification of potential biomarkers and therapeutic targets. FUND: This study was supported by Brain Tumour Research.


Assuntos
Meningioma/metabolismo , Fosfoproteínas/metabolismo , Proteoma , Proteômica , Linhagem Celular Tumoral , Cromatografia Líquida , Biologia Computacional/métodos , Proteínas de Ligação a DNA , Regulação Neoplásica da Expressão Gênica , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Meningioma/genética , Meningioma/patologia , Mutação , Quinases Relacionadas a NIMA/genética , Quinases Relacionadas a NIMA/metabolismo , Fosfopeptídeos/metabolismo , Proteômica/métodos , Proto-Oncogene Mas , Estabilidade de RNA , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Microambiente Tumoral/genética
2.
Mol Biochem Parasitol ; 214: 87-90, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28414172

RESUMO

Successful sporogony of Plasmodium berghei in vector mosquitoes requires expression of a family of six modular proteins named LCCL lectin domain adhesive-like proteins (LAPs). The LAPs share a subcellular localization in the crystalloid, a unique parasite organelle that forms during ookinete development. Here, LAP interactions in P. berghei were studied using a series of parasite lines stably expressing reporter-tagged LAPs combined with affinity purification and high accuracy label free quantitative mass spectrometry. Our results show that abundant complexes containing LAP1, LAP2 and LAP3 are formed in gametocytes through high avidity interactions. Following fertilization, LAP4, LAP5 and LAP6 are recruited to this complex, a process that is facilitated by LAP1 chiefly through its scavenger receptor cysteine-rich modules. These collective findings provide new insight into the temporal and molecular dynamics of protein complex formation that lead up to, and are required for, crystalloid biogenesis and downstream sporozoite transmission of malaria parasites.


Assuntos
Organelas/metabolismo , Plasmodium berghei/metabolismo , Multimerização Proteica , Proteínas de Protozoários/metabolismo , Cromatografia de Afinidade , Espectrometria de Massas , Mapeamento de Interação de Proteínas
3.
EBioMedicine ; 16: 76-86, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28126595

RESUMO

Loss or mutation of the tumour suppressor Merlin predisposes individuals to develop multiple nervous system tumours, including schwannomas and meningiomas, sporadically or as part of the autosomal dominant inherited condition Neurofibromatosis 2 (NF2). These tumours display largely low grade features but their presence can lead to significant morbidity. Surgery and radiotherapy remain the only treatment options despite years of research, therefore an effective therapeutic is required. Unbiased omics studies have become pivotal in the identification of differentially expressed genes and proteins that may act as drug targets or biomarkers. Here we analysed the proteome and phospho-proteome of these genetically defined tumours using primary human tumour cells to identify upregulated/activated proteins and/or pathways. We identified over 2000 proteins in comparative experiments between Merlin-deficient schwannoma and meningioma compared to human Schwann and meningeal cells respectively. Using functional enrichment analysis we highlighted several dysregulated pathways and Gene Ontology terms. We identified several proteins and phospho-proteins that are more highly expressed in tumours compared to controls. Among proteins jointly dysregulated in both tumours we focused in particular on PDZ and LIM domain protein 2 (PDLIM2) and validated its overexpression in several tumour samples, while not detecting it in normal cells. We showed that shRNA mediated knockdown of PDLIM2 in both primary meningioma and schwannoma leads to significant reductions in cellular proliferation. To our knowledge, this is the first comprehensive assessment of the NF2-related meningioma and schwannoma proteome and phospho-proteome. Taken together, our data highlight several commonly deregulated factors, and indicate that PDLIM2 may represent a novel, common target for meningioma and schwannoma.


Assuntos
Proteínas com Domínio LIM/metabolismo , Meningioma/metabolismo , Proteínas dos Microfilamentos/metabolismo , Neurilemoma/metabolismo , Fosfoproteínas/metabolismo , Proteoma/metabolismo , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/genética , Células Cultivadas , Cromatografia Líquida , Humanos , Proteínas com Domínio LIM/genética , Meningioma/genética , Meningioma/patologia , Proteínas dos Microfilamentos/genética , Microscopia Confocal , Neurilemoma/genética , Neurilemoma/patologia , Neurofibromina 2/deficiência , Neurofibromina 2/genética , Fosfoproteínas/genética , Mapas de Interação de Proteínas , Proteoma/genética , Proteômica/métodos , Interferência de RNA , Espectrometria de Massas em Tandem
4.
Eur J Cell Biol ; 95(11): 465-474, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27402209

RESUMO

Neuroblastoma is the second-most common solid tumor in children and originates from poorly differentiated neural crest-derived progenitors. Although most advanced stage metastatic neuroblastoma patients initially respond to treatment, a therapy resistant pool of poorly differentiated cells frequently arises, leading to refractory disease. A lack of insight into the molecular mechanisms that underlie neuroblastoma progression hampers the development of effective new therapies for these patients. Normal neural crest development and maturation is guided by physical interactions between the cell and its surroundings, in addition to soluble factors such as growth factors. This mechanical crosstalk is mediated by actin-based adhesion structures and cell protrusions that probe the cellular environment to modulate migration, proliferation, survival and differentiation. Whereas such signals preserve cellular quiescence in non-malignant cells, perturbed adhesion signaling promotes de-differentiation, uncontrolled cell proliferation, tissue invasion and therapy resistance. We previously reported that high expression levels of the channel-kinase TRPM7, a protein that maintains the progenitor state of embryonic neural crest cells, are closely associated with progenitor-like features of tumor cells, accompanied by extensive cytoskeletal reorganization and adhesion remodeling. To define mechanisms by which TRPM7 may contribute to neuroblastoma progression, we applied a proteomics approach to identify TRPM7 interacting proteins. We show that TRPM7 is part of a large complex of proteins, many of which function in cytoskeletal organization, cell protrusion formation and adhesion dynamics. Expression of a subset of these TRPM7 interacting proteins strongly correlates with neuroblastoma progression in independent neuroblastoma patient datasets. Thus, TRPM7 is part of a large cytoskeletal complex that may affect the malignant potential of tumor cells by regulating actomyosin dynamics and cell-matrix interactions.


Assuntos
Citoesqueleto/metabolismo , Proteínas de Neoplasias/metabolismo , Neuroblastoma/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Canais de Cátion TRPM/metabolismo , Actomiosina/genética , Actomiosina/metabolismo , Animais , Linhagem Celular Tumoral , Citoesqueleto/genética , Citoesqueleto/patologia , Bases de Dados Genéticas , Humanos , Camundongos , Proteínas de Neoplasias/genética , Neuroblastoma/genética , Neuroblastoma/patologia , Proteínas Serina-Treonina Quinases/genética , Canais de Cátion TRPM/genética
5.
Blood Cells Mol Dis ; 58: 35-44, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27067487

RESUMO

The phosphorylation status of red blood cell proteins is strongly altered during the infection by the malaria parasite Plasmodium falciparum. We identify the key phosphorylation events that occur in the erythrocyte membrane and cytoskeleton during infection, by a comparative analysis of global phospho-proteome screens between infected (obtained at schizont stage) and uninfected RBCs. The meta-analysis of reported mass spectrometry studies revealed a novel compendium of 495 phosphorylation sites in 182 human proteins with regulatory roles in red cell morphology and stability, with about 25% of these sites specific to infected cells. A phosphorylation motif analysis detected 7 unique motifs that were largely mapped to kinase consensus sequences of casein kinase II and of protein kinase A/protein kinase C. This analysis highlighted prominent roles for PKA/PKC involving 78 phosphorylation sites. We then compared the phosphorylation status of PKA (PKC) specific sites in adducin, dematin, Band 3 and GLUT-1 in uninfected RBC stimulated or not by cAMP to their phosphorylation status in iRBC. We showed cAMP-induced phosphorylation of adducin S59 by immunoblotting and we were able to demonstrate parasite-induced phosphorylation for adducin S726, Band 3 and GLUT-1, corroborating the protein phosphorylation status in our erythrocyte phosphorylation site compendium.


Assuntos
Eritrócitos/parasitologia , Malária Falciparum/metabolismo , Plasmodium falciparum/fisiologia , Proteoma/metabolismo , Sequência de Aminoácidos , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/análise , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citoesqueleto/química , Citoesqueleto/metabolismo , Citoesqueleto/parasitologia , Eritrócitos/química , Eritrócitos/metabolismo , Transportador de Glucose Tipo 1/análise , Transportador de Glucose Tipo 1/metabolismo , Humanos , Fosforilação , Proteoma/análise
6.
J Proteomics ; 84: 132-47, 2013 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-23584145

RESUMO

SHP2/PTPN11 is a key regulator of cytokine, growth factor and integrin signaling. SHP2 influences cell survival, proliferation and differentiation by regulating major signaling pathways. Mutations in PTPN11 cause severe diseases like Noonan, LEOPARD syndrome or leukemia. Whereas several of these mutations result in altered enzymatic activity due to impaired auto-inhibition, not all disease patterns can be explained by this mechanism. In this study we analyzed altered binding properties of disease-related SHP2-mutants bearing point mutations within the SH2-domain (T42A, E139D, and R138Q). Mutants were chosen according to SPR assays, which revealed different binding properties of mutated SH2 towards phosphorylated receptor peptides. To analyze global changes in mutant binding properties we applied quantitative mass spectrometry (SILAC). Using an in vitro approach we identified overall more than 1000 protein candidates, which specifically bind to the SH2-domain of SHP2. We discovered that mutations in the SH2-domain selectively affected protein enrichment by altering the binding capacity of the SH2-domain. Mutation-dependent, enhanced or reduced exposure of SHP2 to its binding partners could have an impact on the dynamics of signaling networks. Thus, disease-associated mutants of SHP2 should not only be discussed in the context of deregulated auto-inhibition but also with respect to deregulated protein targeting of the SHP2 mutants. BIOLOGICAL SIGNIFICANCE: Using quantitative mass spectrometry based proteomics we provided evidence that disease related mutations in SHP2 domains of SHP2 are able to influence SHP2 recruitment to its targets in mutation dependent manner. We discovered that mutations in the SH2-domain selectively affected protein enrichment ratios suggesting altered binding properties of the SH2-domain. We demonstrated that mutations within SHP2, which had been attributed to affect the enzymatic activity (i.e. affect the open/close status of SHP2), also differ in respect to binding properties. Our study indicates that SHP2 mutations need to be discussed not only in terms of deregulated auto-inhibition but also with respect to deregulated protein targeting properties of the SHP2 mutants. Discovery of the new binding partners for disease-related SHP2 mutants might provide a fruitful foundation for developing strategies targeting Noonan-associated leukemia.


Assuntos
Leucemia/enzimologia , Mutação de Sentido Incorreto , Proteínas de Neoplasias/metabolismo , Síndrome de Noonan/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Substituição de Aminoácidos , Células HeLa , Humanos , Leucemia/genética , Leucemia/patologia , Proteínas de Neoplasias/genética , Síndrome de Noonan/genética , Síndrome de Noonan/patologia , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Domínios de Homologia de src
7.
J Proteome Res ; 11(11): 5323-37, 2012 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-23025827

RESUMO

The asexual blood stages of Plasmodium falciparum cause the most lethal form of human malaria. During growth within an infected red blood cell, parasite multiplication and formation of invasive merozoites is called schizogony. Here, we present a detailed analysis of the phosphoproteome of P. falciparum schizonts revealing 2541 unique phosphorylation sites, including 871 novel sites. Prominent roles for cAMP-dependent protein kinase A- and phosphatidylinositol-signaling were identified following analysis by functional enrichment, phosphoprotein interaction network clustering and phospho-motif identification tools. We observed that most key enzymes in the inositol pathway are phosphorylated, which strongly suggests additional levels of regulation and crosstalk with other protein kinases that coregulate different biological processes. A distinct pattern of phosphorylation of proteins involved in merozoite egress and red blood cell invasion was noted. The analyses also revealed that cAMP-PKA signaling is implicated in a wide variety of processes including motility. We verified this finding experimentally using an in vitro kinase assay and identified three novel PKA substrates associated with the glideosome motor complex: myosin A, GAP45 and CDPK1. Therefore, in addition to an established role for CDPK1 in the motor complex, this study reveals the coinvolvement of PKA, further implicating cAMP as an important regulator of host cell invasion.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fosfatidilinositóis/metabolismo , Fosfoproteínas/metabolismo , Plasmodium falciparum/metabolismo , Proteoma , Proteínas de Protozoários/metabolismo , Transdução de Sinais , Animais , Cromatografia Líquida , Eletroforese em Gel de Poliacrilamida , Espectrometria de Massas em Tandem
8.
J Leukoc Biol ; 87(6): 1029-39, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20181724

RESUMO

DC are professional APCs that initiate and regulate adaptive immune responses by interacting with naïve and memory T cells. Chemokines released by DC play an essential role in T cell recruitment and in the maintenance of antigen-specific T cell-DC conjugates. Here, we characterized the expression of the T cell-attracting chemokine CXCL16 by murine DC. We demonstrate that through alternative RNA splicing, DC not only express the previously characterized transmembrane CXCL16 isoform, which can be cleaved from the cell surface, but also a novel isoform lacking the transmembrane and cytoplasmic domains. Transfection of HEK293 cells shows that this novel isoform, termed CXCL16v, is not expressed on the cell membrane but is secreted as a protein of approximately 10 kDa. Quantitative PCR demonstrates that CXCL16v is broadly expressed in lymphoid and nonlymphoid tissues resembling the tissue distribution of DC. Indeed, CXCL16v mRNA is expressed significantly by spleen DC and BM-DC. Moreover, we show that mature DC have increased CXCL16v mRNA levels and express transmembrane and soluble CXCL16 proteins. Finally, we show that CXCL16v specifically attracts cells expressing the chemokine receptor CXCR6. Our data demonstrate that mature DC express secreted, transmembrane, and cleaved CXCL16 isoforms to recruit and communicate efficiently with CXCR6(+) lymphoid cells.


Assuntos
Processamento Alternativo , Quimiocina CXCL6/genética , Fatores Quimiotáticos/metabolismo , Células Dendríticas/metabolismo , Regulação da Expressão Gênica/fisiologia , Receptores CXCR/genética , Animais , Sequência de Bases , Células Cultivadas , Quimiocina CXCL16 , Cromatografia Líquida , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Immunoblotting , Técnicas Imunoenzimáticas , Rim/citologia , Rim/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Isoformas de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores CXCR6 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Espectrometria de Massas em Tandem
9.
J Mol Biol ; 378(4): 790-803, 2008 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-18394644

RESUMO

Deregulation of myosin II-based contractility contributes to the pathogenesis of human diseases, such as cancer, which underscores the necessity for tight spatial and temporal control of myosin II activity. Recently, we demonstrated that activation of the mammalian alpha-kinase TRPM7 inhibits myosin II-based contractility in a Ca(2+)- and kinase-dependent manner. However, the molecular mechanism is poorly defined. Here, we demonstrate that TRPM7 phosphorylates the COOH-termini of both mouse and human myosin IIA heavy chains--the COOH-terminus being a region that is critical for filament stability. Phosphorylated residues were mapped to Thr1800, Ser1803 and Ser1808. Mutation of these residues to alanine and that to aspartic acid lead to an increase and a decrease, respectively, in myosin IIA incorporation into the actomyosin cytoskeleton and accordingly affect subcellular localization. In conclusion, our data demonstrate that TRPM7 regulates myosin IIA filament stability and localization by phosphorylating a short stretch of amino acids within the alpha-helical tail of the myosin IIA heavy chain.


Assuntos
Cadeias Pesadas de Miosina/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Canais de Cátion TRPM/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Sequência Conservada , Humanos , Cinética , Camundongos , Dados de Sequência Molecular , Mutação/genética , Miosina não Muscular Tipo IIA/química , Miosina não Muscular Tipo IIA/genética , Fosforilação , Fosfosserina/metabolismo , Fosfotreonina/metabolismo , Alinhamento de Sequência , Canais de Cátion TRPM/genética
10.
J Cell Biol ; 180(5): 915-29, 2008 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-18332217

RESUMO

Cyclin-dependent kinases (Cdks) fulfill key functions in many cellular processes, including cell cycle progression and cytoskeletal dynamics. A limited number of Cdk substrates have been identified with few demonstrated to be regulated by Cdk-dependent phosphorylation. We identify on protein expression arrays novel cyclin E-Cdk2 substrates, including SIRT2, a member of the Sirtuin family of NAD(+)-dependent deacetylases that targets alpha-tubulin. We define Ser-331 as the site phosphorylated by cyclin E-Cdk2, cyclin A-Cdk2, and p35-Cdk5 both in vitro and in cells. Importantly, phosphorylation at Ser-331 inhibits the catalytic activity of SIRT2. Gain- and loss-of-function studies demonstrate that SIRT2 interfered with cell adhesion and cell migration. In postmitotic hippocampal neurons, neurite outgrowth and growth cone collapse are inhibited by SIRT2. The effects provoked by SIRT2, but not those of a nonphosphorylatable mutant, are antagonized by Cdk-dependent phosphorylation. Collectively, our findings identify a posttranslational mechanism that controls SIRT2 function, and they provide evidence for a novel regulatory circuitry involving Cdks, SIRT2, and microtubules.


Assuntos
Movimento Celular/genética , Quinases Ciclina-Dependentes/metabolismo , Sirtuínas/metabolismo , Sequência de Aminoácidos , Animais , Domínio Catalítico , Diferenciação Celular/genética , Linhagem Celular , Ciclina A/genética , Ciclina A/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Quinase 5 Dependente de Ciclina/genética , Quinase 5 Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/genética , Cones de Crescimento/metabolismo , Cones de Crescimento/ultraestrutura , Células HeLa , Hipocampo/embriologia , Hipocampo/metabolismo , Hipocampo/ultraestrutura , Humanos , Camundongos , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional/genética , Serina/metabolismo , Sirtuína 2 , Sirtuínas/genética
11.
J Biol Chem ; 282(23): 17148-56, 2007 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-17426022

RESUMO

Pfs48/45, a member of a Plasmodium-specific protein family, displays conformation-dependent epitopes and is an important target for malaria transmission-blocking (TB) immunity. To design a recombinant Pfs48/45-based TB vaccine, we analyzed the conformational TB epitopes of Pfs48/45. The Pfs48/45 protein was found to consist of a C-terminal six-cysteine module recognized by anti-epitope I antibodies, a middle four-cysteine module recognized by anti-epitopes IIb and III, and an N-terminal module recognized by anti-epitope V antibodies. Refolding assays identified that a fragment of 10 cysteines (10C), comprising the middle four-cysteine and the C-terminal six-cysteine modules, possesses superior refolding capacity. The refolded and partially purified 10C conformer elicited antibodies in mice that targeted at least two of the TB epitopes (I and III). The induced antibodies could block the fertilization of Plasmodium falciparum gametes in vivo in a concentration-dependent manner. Our results provide important insight into the structural organization of the Pfs48/45 protein and experimental support for a Pfs48/45-based subunit vaccine.


Assuntos
Anticorpos Antiprotozoários/biossíntese , Antígenos de Protozoários/imunologia , Epitopos/imunologia , Glicoproteínas de Membrana/imunologia , Proteínas de Protozoários/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/química , Ensaio de Imunoadsorção Enzimática , Epitopos/química , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
12.
Int J Cancer ; 119(9): 2127-35, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16841330

RESUMO

The human bowel contains a large and dynamic bacterial population that is not only essential for intestinal health, but also critical for the development of diseases such as cancer. In this respect, the Gram-positive bacterium Streptococcus bovis has been associated with colon cancer for many years. To investigate the clinical importance of this association, an immunocapture mass spectrometry assay was developed that can generate infection-related protein profiles. The composition of these profiles is governed by the capture of specific antigens by serum antibodies from colon cancer patients. This assay showed that S. bovis antigen profiles could distinguish 11 out of 12 colon cancer patients from 8 control subjects, whereas antigen profiles derived from the gut bacterium Escherichia coli were not diagnostic for colon cancer. Moreover, S. bovis antigen profiles were also detected in polyp patients, indicating that infection with this bacterium does occur early during carcinogenesis. Highly accurate tandem mass spectrometry was used to identify one of the diagnostic antigens as a surface-exposed heparin-binding protein, which might be involved in attachment of S. bovis to tumor cells. Together, these findings corroborate the hypothesis that colonic lesions provide a specific niche for S. bovis, resulting in tumor-associated "silent" infections. These infections, however, only become apparent in colon cancer patients with a compromised immune system (bacteremia) or coincidental cardiac valve lesions (endocarditis). This makes profiling of the humoral immune response against "silent" S. bovis infections a promising diagnostic tool for the early detection of human colon cancer, which is crucial for the effective treatment of this disease.


Assuntos
Formação de Anticorpos , Antígenos de Bactérias/sangue , Linfócitos B/imunologia , Neoplasias do Colo/microbiologia , Streptococcus bovis/imunologia , Neoplasias do Colo/diagnóstico , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Humanos , Estadiamento de Neoplasias , Streptococcus bovis/isolamento & purificação
13.
Mol Cell Biol ; 26(3): 843-51, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16428440

RESUMO

The human genome contains a number of methyl CpG binding proteins that translate DNA methylation into a physiological response. To gain insight into the function of MBD2 and MBD3, we first applied protein tagging and mass spectrometry. We show that MBD2 and MBD3 assemble into mutually exclusive distinct Mi-2/NuRD-like complexes, called MBD2/NuRD and MBD3/NuRD. We identified DOC-1, a putative tumor suppressor, as a novel core subunit of MBD2/NuRD as well as MBD3/NuRD. PRMT5 and its cofactor MEP50 were identified as specific MBD2/NuRD interactors. PRMT5 stably and specifically associates with and methylates the RG-rich N terminus of MBD2. Chromatin immunoprecipitation experiments revealed that PRMT5 and MBD2 are recruited to CpG islands in a methylation-dependent manner in vivo and that H4R3, a substrate of PRMT, is methylated at these loci. Our data show that MBD2/NuRD and MBD3/NuRD are distinct protein complexes with different biochemical and functional properties.


Assuntos
Metilação de DNA , Proteínas de Ligação a DNA/metabolismo , Histona Desacetilases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Bases , Células Cultivadas , Cromatina/metabolismo , Citocinas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase , Dados de Sequência Molecular , Proteínas Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA