Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
2.
J Biol Chem ; 291(42): 21893-21902, 2016 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-27573245

RESUMO

The synthesis, processing, and joining of Okazaki fragments during DNA replication is complex, requiring the sequential action of a large number of proteins. Proliferating cell nuclear antigen, a DNA sliding clamp, interacts with and coordinates the activity of several DNA replication proteins, including the enzymes flap endonuclease 1 (FEN-1) and DNA ligase I that complete the processing and joining of Okazaki fragments, respectively. Although it is evident that maintaining the appropriate relative stoichiometry of FEN-1 and DNA ligase I, which compete for binding to proliferating cell nuclear antigen, is critical to prevent genomic instability, little is known about how the steady state levels of DNA replication proteins are regulated, in particular the proteolytic mechanisms involved in their turnover. Because DNA ligase I has been reported to be ubiquitylated, we used a proteomic approach to map ubiquitylation sites and screen for DNA ligase I-associated E3 ubiquitin ligases. We identified three ubiquitylated lysine residues and showed that DNA ligase I interacts with and is targeted for ubiquitylation by DCAF7, a specificity factor for the Cul4-DDB1 complex. Notably, knockdown of DCAF7 reduced the degradation of DNA ligase I in response to inhibition of proliferation and replacement of ubiquitylated lysine residues reduced the in vitro ubiquitylation of DNA ligase I by Cul4-DDB1 and DCAF7. In contrast, a different E3 ubiquitin ligase regulates FEN-1 turnover. Thus, although the expression of many of the genes encoding DNA replication proteins is coordinately regulated, our studies reveal that different mechanisms are involved in the turnover of these proteins.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Culina/metabolismo , DNA Ligase Dependente de ATP/metabolismo , Proteínas de Ligação a DNA/metabolismo , Complexos Multienzimáticos/metabolismo , Proteólise , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular , Proliferação de Células/fisiologia , Proteínas Culina/genética , DNA Ligase Dependente de ATP/genética , Proteínas de Ligação a DNA/genética , Feminino , Endonucleases Flap/genética , Endonucleases Flap/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Complexos Multienzimáticos/genética , Ubiquitinação/fisiologia
3.
Anal Chem ; 87(20): 10462-9, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26378940

RESUMO

Exosomes are microvesicles of endocytic origin constitutively released by multiple cell types into the extracellular environment. With evidence that exosomes can be detected in the blood of patients with various malignancies, the development of a platform that uses exosomes as a diagnostic tool has been proposed. However, it has been difficult to truly define the exosome proteome due to the challenge of discerning contaminant proteins that may be identified via mass spectrometry using various exosome enrichment strategies. To better define the exosome proteome in breast cancer, we incorporated a combination of Tandem-Mass-Tag (TMT) quantitative proteomics approach and Support Vector Machine (SVM) cluster analysis of three conditioned media derived fractions corresponding to a 10 000g cellular debris pellet, a 100 000g crude exosome pellet, and an Optiprep enriched exosome pellet. The quantitative analysis identified 2 179 proteins in all three fractions, with known exosomal cargo proteins displaying at least a 2-fold enrichment in the exosome fraction based on the TMT protein ratios. Employing SVM cluster analysis allowed for the classification 251 proteins as "true" exosomal cargo proteins. This study provides a robust and vigorous framework for the future development of using exosomes as a potential multiprotein marker phenotyping tool that could be useful in breast cancer diagnosis and monitoring disease progression.


Assuntos
Neoplasias da Mama/metabolismo , Exossomos/química , Proteoma/análise , Proteômica , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/patologia , Análise por Conglomerados , Exossomos/metabolismo , Feminino , Humanos , Análise Multivariada , Espectrometria de Massas em Tandem , Células Tumorais Cultivadas
4.
Biochem J ; 462(1): 77-88, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24869773

RESUMO

In Alzheimer's disease, the microtubule-associated protein tau dissociates from the neuronal cytoskeleton and aggregates to form cytoplasmic inclusions. Although hyperphosphorylation of tau serine and threonine residues is an established trigger of tau misfunction and aggregation, tau modifications extend to lysine residues as well, raising the possibility that different modification signatures depress or promote aggregation propensity depending on site occupancy. To identify lysine residue modifications associated with normal tau function, soluble tau proteins isolated from four cognitively normal human brains were characterized by MS methods. The major detectable lysine modification was found to be methylation, which appeared in the form of mono- and di-methyl lysine residues distributed among at least 11 sites. Unlike tau phosphorylation sites, the frequency of lysine methylation was highest in the microtubule-binding repeat region that mediates both microtubule binding and homotypic interactions. When purified recombinant human tau was modified in vitro through reductive methylation, its ability to promote tubulin polymerization was retained, whereas its aggregation propensity was greatly attenuated at both nucleation and extension steps. These data establish lysine methylation as part of the normal tau post-translational modification signature in human brain, and suggest that it can function in part to protect against pathological tau aggregation.


Assuntos
Lisina/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas tau/metabolismo , Sequência de Aminoácidos , Humanos , Masculino , Metilação , Microtúbulos/metabolismo , Pessoa de Meia-Idade , Fosforilação , Estrutura Quaternária de Proteína , Espectrometria de Massas em Tandem , Tubulina (Proteína)/metabolismo
5.
J Biol Chem ; 287(44): 36711-9, 2012 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-22952233

RESUMO

Human DNA ligase I (hLigI) joins Okazaki fragments during DNA replication and completes excision repair via interactions with proliferating cell nuclear antigen and replication factor C (RFC). Unlike proliferating cell nuclear antigen, the interaction with RFC is regulated by hLigI phosphorylation. To identity of the site(s) involved in this regulation, we analyzed phosphorylated hLigI purified from insect cells by mass spectrometry. These results suggested that serine 51 phosphorylation negatively regulates the interaction with RFC. Therefore, we constructed versions of hLigI in which serine 51 was replaced with either alanine (hLigI51A) to prevent phosphorylation or aspartic acid (hLigI51D) to mimic phosphorylation. hLigI51D but not hLigI51A was defective in binding to purified RFC and in associating with RFC in cell extracts. Although DNA synthesis and proliferation of hLigI-deficient cells expressing either hLig51A or hLig51 was reduced compared with cells expressing wild-type hLigI, cellular senescence was only observed in the cells expressing hLigI51D. Notably, these cells had increased levels of spontaneous DNA damage and phosphorylated CHK2. In addition, although expression of hLigI51A complemented the sensitivity of hLigI-deficient cells to a poly (ADP-ribose polymerase (PARP) inhibitor, expression of hLig151D did not, presumably because these cells are more dependent upon PARP-dependent repair pathways to repair the damage resulting from the abnormal DNA replication. Finally, neither expression of hLigI51D nor hLigI51A fully complemented the sensitivity of hLigI-deficient cells to DNA alkylation. Thus, phosphorylation of serine 51 on hLigI plays a critical role in regulating the interaction between hLigI and RFC, which is required for efficient DNA replication and repair.


Assuntos
DNA Ligases/metabolismo , Reparo do DNA , Replicação do DNA , Processamento de Proteína Pós-Traducional , Proteína de Replicação C/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Linhagem Celular , Proliferação de Células , Forma Celular , Senescência Celular , Dano ao DNA , DNA Ligase Dependente de ATP , DNA Ligases/química , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Mapeamento de Peptídeos , Fosforilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Proteína de Replicação C/química , Serina/metabolismo
6.
Anal Chem ; 84(10): 4535-43, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22519468

RESUMO

Accurate protein identification and quantitation are critical when interpreting the biological relevance of large-scale shotgun proteomics data sets. Although significant technical advances in peptide and protein identification have been made, accurate quantitation of high-throughput data sets remains a key challenge in mass spectrometry data analysis and is a labor intensive process for many proteomics laboratories. Here, we report a new SILAC-based proteomics quantitation software tool, named IsoQuant, which is used to process high mass accuracy mass spectrometry data. IsoQuant offers a convenient quantitation framework to calculate peptide/protein relative abundance ratios. At the same time, it also includes a visualization platform that permits users to validate the quality of SILAC peptide and protein ratios. The program is written in the C# programming language under the Microsoft .NET framework version 4.0 and has been tested to be compatible with both 32-bit and 64-bit Windows 7. It is freely available to noncommercial users at http://www.proteomeumb.org/MZw.html .


Assuntos
Aminoácidos/química , Proteínas/análise , Software , Espectrometria de Massas em Tandem , Animais , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Bases de Dados de Proteínas , Hipocampo/metabolismo , Marcação por Isótopo , Proteômica , Ratos
7.
Acta Neuropathol ; 123(1): 105-17, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22033876

RESUMO

In sporadic Alzheimer's disease (AD), neurofibrillary lesion formation is preceded by extensive post-translational modification of the microtubule associated protein tau. To identify the modification signature associated with tau lesion formation at single amino acid resolution, immunopurified paired helical filaments were isolated from AD brain and subjected to nanoflow liquid chromatography-tandem mass spectrometry analysis. The resulting spectra identified monomethylation of lysine residues as a new tau modification. The methyl-lysine was distributed among seven residues located in the projection and microtubule binding repeat regions of tau protein, with one site, K254, being a substrate for a competing lysine modification, ubiquitylation. To characterize methyl lysine content in intact tissue, hippocampal sections prepared from post mortem late-stage AD cases were subjected to double-label confocal fluorescence microscopy using anti-tau and anti-methyl lysine antibodies. Anti-methyl lysine immunoreactivity colocalized with 78 ± 13% of neurofibrillary tangles in these specimens. Together these data provide the first evidence that tau in neurofibrillary lesions is post-translationally modified by lysine methylation.


Assuntos
Doença de Alzheimer/metabolismo , Lisina/metabolismo , Ubiquitinação/fisiologia , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Feminino , Humanos , Lisina/química , Masculino , Metilação , Pessoa de Meia-Idade , Dados de Sequência Molecular , Emaranhados Neurofibrilares/metabolismo , Emaranhados Neurofibrilares/patologia , Fosforilação/fisiologia , Processamento de Proteína Pós-Traducional , Espectrometria de Massas em Tandem/métodos , Proteínas tau/química
8.
Brain ; 134(Pt 1): 258-77, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21186265

RESUMO

Autophagy, a major degradative pathway for proteins and organelles, is essential for survival of mature neurons. Extensive autophagic-lysosomal pathology in Alzheimer's disease brain contributes to Alzheimer's disease pathogenesis, although the underlying mechanisms are not well understood. Here, we identified and characterized marked intraneuronal amyloid-ß peptide/amyloid and lysosomal system pathology in the Alzheimer's disease mouse model TgCRND8 similar to that previously described in Alzheimer's disease brains. We further establish that the basis for these pathologies involves defective proteolytic clearance of neuronal autophagic substrates including amyloid-ß peptide. To establish the pathogenic significance of these abnormalities, we enhanced lysosomal cathepsin activities and rates of autophagic protein turnover in TgCRND8 mice by genetically deleting cystatin B, an endogenous inhibitor of lysosomal cysteine proteases. Cystatin B deletion rescued autophagic-lysosomal pathology, reduced abnormal accumulations of amyloid-ß peptide, ubiquitinated proteins and other autophagic substrates within autolysosomes/lysosomes and reduced intraneuronal amyloid-ß peptide. The amelioration of lysosomal function in TgCRND8 markedly decreased extracellular amyloid deposition and total brain amyloid-ß peptide 40 and 42 levels, and prevented the development of deficits of learning and memory in fear conditioning and olfactory habituation tests. Our findings support the pathogenic significance of autophagic-lysosomal dysfunction in Alzheimer's disease and indicate the potential value of restoring normal autophagy as an innovative therapeutic strategy for Alzheimer's disease.


Assuntos
Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Autofagia/fisiologia , Encéfalo/patologia , Transtornos da Memória/fisiopatologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Animais , Western Blotting , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Condicionamento Psicológico , Ensaio de Imunoadsorção Enzimática , Medo , Habituação Psicofisiológica , Imuno-Histoquímica , Lisossomos/metabolismo , Lisossomos/patologia , Transtornos da Memória/genética , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Neurônios/patologia
9.
Nature ; 440(7082): 352-7, 2006 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-16541076

RESUMO

Memory function often declines with age, and is believed to deteriorate initially because of changes in synaptic function rather than loss of neurons. Some individuals then go on to develop Alzheimer's disease with neurodegeneration. Here we use Tg2576 mice, which express a human amyloid-beta precursor protein (APP) variant linked to Alzheimer's disease, to investigate the cause of memory decline in the absence of neurodegeneration or amyloid-beta protein amyloidosis. Young Tg2576 mice (< 6 months old) have normal memory and lack neuropathology, middle-aged mice (6-14 months old) develop memory deficits without neuronal loss, and old mice (> 14 months old) form abundant neuritic plaques containing amyloid-beta (refs 3-6). We found that memory deficits in middle-aged Tg2576 mice are caused by the extracellular accumulation of a 56-kDa soluble amyloid-beta assembly, which we term Abeta*56 (Abeta star 56). Abeta*56 purified from the brains of impaired Tg2576 mice disrupts memory when administered to young rats. We propose that Abeta*56 impairs memory independently of plaques or neuronal loss, and may contribute to cognitive deficits associated with Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/metabolismo , Encéfalo/fisiopatologia , Memória/fisiologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/farmacologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Memória/efeitos dos fármacos , Camundongos , Peso Molecular , Placa Amiloide/química , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Estrutura Quaternária de Proteína , Ratos , Solubilidade , Fatores de Tempo
10.
Neuron ; 110(8): 1304-1317, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35298921

RESUMO

The ε4 allele of the apolipoprotein E gene (APOE4) is a strong genetic risk factor for Alzheimer's disease (AD) and several other neurodegenerative conditions, including Lewy body dementia (LBD). The three APOE alleles encode protein isoforms that differ from one another only at amino acid positions 112 and 158: apoE2 (C112, C158), apoE3 (C112, R158), and apoE4 (R112, R158). Despite progress, it remains unclear how these small amino acid differences in apoE sequence among the three isoforms lead to profound effects on aging and disease-related pathways. Here, we propose a novel "ApoE Cascade Hypothesis" in AD and age-related cognitive decline, which states that the biochemical and biophysical properties of apoE impact a cascade of events at the cellular and systems levels, ultimately impacting aging-related pathogenic conditions including AD. As such, apoE-targeted therapeutic interventions are predicted to be more effective by addressing the biochemical phase of the cascade.


Assuntos
Doença de Alzheimer , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Aminoácidos , Apolipoproteína E2/genética , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Apolipoproteínas E/metabolismo , Humanos , Isoformas de Proteínas/metabolismo
11.
Anal Chem ; 83(14): 5511-8, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21619060

RESUMO

The analysis of protein interactors in protein complexes can yield important insight into protein function and signal transduction. Thus, a reliable approach to distinguish true interactors from nonspecific interacting proteins is of utmost importance for accurate data interpretation. Although stringent purification methods are critical, challenges still remain in the selection of criteria that will permit the objective differentiation of true members of the protein complex from nonspecific background proteins. To address these challenges, we have developed a quantitative proteomic strategy combining stable isotope labeling with amino acids in cell culture (SILAC), affinity substrate trapping, and gel electrophoresis followed by liquid chromatography-tandem mass spectrometry (geLC-MS/MS) protein quantitation. ATP hydrolysis-deficient vacuolar protein sorting-associated protein 4B (Vps4B) was used as the "bait" protein which served as a substrate trap since its lack of ATP hydrolysis enzymatic activity allows the stabilization of its transiently associated interacting proteins. A significant advantage of our approach is the use of our new in-house-developed software program for SILAC-based mass spectrometry quantitation, which further facilitates the differentiation between the bait protein, endogenous bait-interacting proteins, and nonspecific binding proteins based on their protein ratios. The strategy presented herein is applicable to the analysis of other protein complexes whose compositions are dependent upon the ATP hydrolysis activity of the bait protein used in affinity purification studies.


Assuntos
Adenosina Trifosfatases/metabolismo , Aminoácidos/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Marcação por Isótopo/métodos , Mapeamento de Interação de Proteínas/métodos , Espectrometria de Massas em Tandem/métodos , ATPases Associadas a Diversas Atividades Celulares , Antibacterianos/farmacologia , Células HEK293 , Humanos , Proteômica/métodos , Tetraciclina/farmacologia
12.
World J Gastroenterol ; 27(22): 2944-2962, 2021 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-34168400

RESUMO

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 is a global pandemic and poses a major threat to human health worldwide. In addition to respiratory symptoms, COVID-19 is usually accompanied by systemic inflammation and liver damage in moderate and severe cases. Nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor that regulates the expression of antioxidant proteins, participating in COVID-19-mediated inflammation and liver injury. Here, we show the novel reciprocal regulation between NRF2 and inflammatory mediators associated with COVID-19-related liver injury. Additionally, we describe some mechanisms and treatment strategies.


Assuntos
COVID-19 , Mediadores da Inflamação , Hepatopatias/virologia , Fator 2 Relacionado a NF-E2 , COVID-19/patologia , Humanos , Mediadores da Inflamação/metabolismo , Fígado/metabolismo , Fígado/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , SARS-CoV-2 , Transdução de Sinais
13.
Cell Death Dis ; 11(4): 214, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32238799

RESUMO

The DNA damage response (DDR) pathway is a promising target for anticancer therapies. The androgen receptor and myeloblastosis transcription factors have been reported to regulate expression of an overlapping set of DDR genes in prostate cancer cells. Here, we found that histone demethylase JMJD1A regulates expression of a different set of DDR genes largely through c-Myc. Inhibition of JMJD1A delayed the resolution of γ-H2AX foci, reduced the formation of foci containing ubiquitin, 53BP1, BRCA1 or Rad51, and inhibited the reporter activity of double-strand break (DSB) repair. Mechanistically, JMJD1A regulated expression of DDR genes by increasing not only the level but also the chromatin recruitment of c-Myc through H3K9 demethylation. Further, we found that ubiquitin ligase HUWE1 induced the K27-/K29-linked noncanonical ubiquitination of JMJD1A at lysine-918. Ablation of the JMJD1A noncanonical ubiquitination lowered DDR gene expression, impaired DSB repair, and sensitized response of prostate cells to irradiation, topoisomerase inhibitors or PARP inhibitors. Thus, development of agents that target JMJD1A or its noncanonical ubiquitination may sensitize the response of prostate cancer to radiotherapy and possibly also genotoxic therapy.


Assuntos
Reparo do DNA , Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias de Próstata Resistentes à Castração/radioterapia , Animais , Quebras de DNA de Cadeia Dupla , Modelos Animais de Doenças , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Camundongos , Células PC-3 , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/metabolismo , Rad51 Recombinase/metabolismo , Tolerância a Radiação , Distribuição Aleatória , Transfecção , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Methods Mol Biol ; 566: 109-21, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20058168

RESUMO

Posttranslational modifications such as phosphorylation and ubiquitination serve, independently or together, as gatekeepers of protein transport and turnover in normal and disease physiologies. Aberrant protein phosphorylation is one of the defining pathological hallmarks of more than 20 different neurodegenerative disorders, including Alzheimer's disease (AD). The disruption of the phosphorylation of neurotransmitter receptors has been implicated as one of the causal factors of impaired memory function in AD (1-3). Another feature of AD is the aberrant accumulation of proteins that are normally degraded by the ubiquitin proteasome system upon being conjugated to ubiquitin. Thus, elucidating the protein targets of phosphorylation and ubiquitination that can serve as AD biomarkers will aid in the development of effective therapeutic approaches to the treatment of AD. This chapter provides details pertaining to the qualitative and quantitative liquid chromatography tandem mass spectrometry-based analysis of an affinity purified, phosphorylated, and ubiquitinated protein, paired-helical filament tau.


Assuntos
Doença de Alzheimer/fisiopatologia , Processamento de Proteína Pós-Traducional , Proteoma/análise , Proteínas tau , Doença de Alzheimer/patologia , Cromatografia Líquida/métodos , Eletroforese em Gel de Poliacrilamida/métodos , Humanos , Espectrometria de Massas/métodos , Fosforilação , Ubiquitinação , Proteínas tau/química , Proteínas tau/metabolismo
15.
J Alzheimers Dis ; 71(3): 979-991, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31450505

RESUMO

Tau is a microtubule-associated protein that normally interacts in monomeric form with the neuronal cytoskeleton. In Alzheimer's disease, however, it aggregates to form the structural component of neurofibrillary lesions. The transformation is controlled in part by age- and disease-associated post-translational modifications. Recently we reported that tau isolated from cognitively normal human brain was methylated on lysine residues, and that high-stoichiometry methylation depressed tau aggregation propensity in vitro. However, whether methylation stoichiometry reached levels needed to influence aggregation propensity in human brain was unknown. Here we address this problem using liquid chromatography-tandem mass spectrometry approaches and human-derived tau samples. Results revealed that lysine methylation was present in soluble tau isolated from cognitively normal elderly cases at multiple sites that only partially overlapped with the distributions reported for cognitively normal middle aged and AD cohorts, and that the quality of methylation shifted from predominantly dimethyl-lysine to monomethyl-lysine with aging and disease. However, bulk mol methylation/mol tau stoichiometries never exceeded 1 mol methyl group/mol tau protein. We conclude that lysine methylation is a physiological post-translational modification of tau protein that changes qualitatively with aging and disease, and that pharmacological elevation of tau methylation may provide a means for protecting against pathological tau aggregation.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Lisina/metabolismo , Proteínas tau/metabolismo , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Feminino , Humanos , Masculino , Metabolômica , Metilação , Pessoa de Meia-Idade , Fosforilação , Processamento de Proteína Pós-Traducional , Proteômica , Espectrometria de Massas em Tandem , Proteínas tau/química
16.
Invest Ophthalmol Vis Sci ; 48(4): 1447-57, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17389470

RESUMO

PURPOSE: To increase knowledge of the biochemical composition of lenticular exfoliation material (XFM) by using proteomic approaches. METHODS: Anterior lens capsules from patients with and without exfoliation syndrome (XFS) were homogenized in formic acid and subjected to cyanogen bromide (CNBr) cleavage, and the pattern of chemically generated fragments was compared by SDS-PAGE after silver staining. Unique XFS bands not present in control cases were excised, digested with TPCK-trypsin, and the resultant peptides sequenced with quadrupole time-of-flight mass spectrometry (MS). In parallel experiments, CNBr-fragmented XFM was separately digested in solution with trypsin and elastase, and the resultant peptide mixture was analyzed by liquid chromatography coupled to tandem MS followed by identification through homology searches at nonredundant protein databases. Immunolocalization of the MS-identified components were performed in XFS versus control samples by using conventional immunohistochemical methods and light microscopy. RESULTS: In addition to fibrillin-1, fibronectin, vitronectin, laminin, and amyloid P-component, which are well-known extracellular matrix and basement membrane components of XFM, the proteomic approaches identified the multifunctional protein clusterin and tissue inhibitor of metalloprotease (TIMP)-3 as well as novel molecules, among them fibulin-2, desmocollin-2, the glycosaminoglycans syndecan-3, and versican, membrane metalloproteases of the ADAM family (a disintegrin and metalloprotease), and the initiation component of the classic complement activation pathway C1q. In all cases, classic immunohistochemistry confirmed their location in XFM. CONCLUSIONS: A novel solubilization strategy combined with sensitive proteomic analysis emphasizes the complexity of the XFS deposits and opens new avenues to study the molecular mechanisms involved in the pathogenesis and progression of XFS.


Assuntos
Moléculas de Adesão Celular/metabolismo , Síndrome de Exfoliação/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Proteínas do Olho/metabolismo , Cápsula do Cristalino/metabolismo , Proteínas de Membrana/metabolismo , Proteoma/metabolismo , Idoso , Idoso de 80 Anos ou mais , Extração de Catarata , Cromatografia Líquida , Eletroforese em Gel de Poliacrilamida , Humanos , Técnicas Imunoenzimáticas , Pessoa de Meia-Idade , Proteômica , Espectrometria de Massas em Tandem
17.
Expert Rev Proteomics ; 4(4): 573-83, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17705714

RESUMO

Healthful physiology can be distinguished from unhealthful physiology by focusing upon how a given signal transduction pathway is shifted as a function of disease. In order to distinguish between pathways that contribute to normal versus disease biology, it is necessary to identify components that comprise a protein module. The development of methods that target such differences is essential for the identification, development and validation of biomarkers that can improve the quality of diagnoses and treatment of disease. This review discusses the use of proteomic methods that integrate cell biology, mass spectrometry and bioinformatics, in relation to the analyses of protein signaling modules that are subject to differential phosphorylation. We examine how these methods can be used to distinguish abnormal from normal physiology.


Assuntos
Proteínas/metabolismo , Proteômica , Transdução de Sinais , Humanos , Espectrometria de Massas , Fosforilação
18.
Methods Mol Biol ; 1523: 161-177, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27975250

RESUMO

Recent advances in mass spectrometry (MS)-based proteomics have greatly facilitated the robust identification and quantification of posttranslational modifications (PTMs), including those that are present at substoichiometric site occupancies. The abnormal posttranslational modification and accumulation of the microtubule-associated protein tau has been implicated in the pathogenesis of Alzheimer's disease (AD), and it is thought that the primary mode of regulation of tau occurs through PTMs. Several studies have been published regarding tau phosphorylation; however, other tau PTMs such as ubiquitylation, acetylation, methylation, oxidation, sumoylation, nitration, and glycosylation have not been analyzed as extensively. The comprehensive detection and delineation of these PTMs is critical for drug target discovery and validation. Lysine-directed PTMs including ubiquitylation, acetylation, and methylation play key regulatory roles with respect to the rates of tau turnover and aggregation. MS-based analytical approaches have been used to gain insight into the tau lysine-directed PTM signature that is most closely associated with neurofibrillary lesion formation. This chapter provides details pertaining to the liquid chromatography tandem mass spectrometry (LC-MS/MS)-based analysis of the lysine-directed posttranslational modification of tau.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Espectrometria de Massas/métodos , Proteínas tau/química , Acetilação , Animais , Humanos , Processamento de Proteína Pós-Traducional , Espectrometria de Massas em Tandem , Ubiquitinação
19.
J Proteomics ; 133: 161-169, 2016 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-26739763

RESUMO

Exosomes are 30-100 nm sized membrane vesicles released by cells into the extracellular space that mediate intercellular communication via transfer of proteins and other biological molecules. To better understand the role of these microvesicles in lung carcinogenesis, we employed a Triple SILAC quantitative proteomic strategy to examine the differential protein abundance between exosomes derived from an immortalized normal bronchial epithelial cell line and two non-small cell lung cancer (NSCLC) cell lines harboring distinct activating mutations in the cell signaling molecules: Kirsten rat sarcoma viral oncogene homolog (KRAS) or epidermal growth factor receptor (EGFR). In total, we were able to quantify 721 exosomal proteins derived from the three cell lines. Proteins associated with signal transduction, including EGFR, GRB2 and SRC, were enriched in NSCLC exosomes, and could actively regulate cell proliferation in recipient cells. This study's investigation of the NSCLC exosomal proteome has identified enriched protein cargo that can contribute to lung cancer progression, which may have potential clinical implications in biomarker development for patients with NSCLC. BIOLOGICAL SIGNIFICANCE: The high mortality associated with lung cancer is a result of late-stage diagnosis of the disease. Current screening techniques used for early detection of lung cancer lack the specificity for accurate diagnosis. Exosomes are nano-sized extracellular vesicles, and the increased abundance of select protein cargo in exosomes derived from cancer cells may be used for diagnostic purposes. In this paper, we applied quantitative proteomic analysis to elucidate abundance differences in exosomal protein cargo between two NSCLC cell lines with distinctive oncogene mutations and an immortalized normal bronchial epithelial cell line. This study revealed proteins associated with cell adhesion, the extracellular matrix, and a variety of signaling molecules were enriched in NSCLC exosomes. The present data reveals a protein profile associated with NSCLC exosomes that suggests a role these vesicles have in the progression of lung carcinogenesis, as well as identifies several promising candidates that could be utilized as a multi-marker protein panel in a diagnostic platform for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Exossomos/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Proteômica , Animais , Linhagem Celular Tumoral , Humanos , Ratos
20.
Theranostics ; 6(1): 65-77, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26722374

RESUMO

Protein glycosylation plays a fundamental role in a multitude of biological processes, and the associated aberrant expression of glycoproteins in cancer has made them attractive biomarkers and therapeutic targets. In this study, we examined differentially expressed glycoproteins in cell lines derived from three different states of lung tumorigenesis: an immortalized bronchial epithelial cell (HBE) line, a non-small cell lung cancer (NSCLC) cell line harboring a Kirsten rat sarcoma viral oncogene homolog (KRAS) activation mutation and a NSCLC cell line harboring an epidermal growth factor receptor (EGFR) activation deletion. Using a Triple SILAC proteomic quantification strategy paired with hydrazide chemistry N-linked glycopeptide enrichment, we quantified 118 glycopeptides in the three cell lines derived from 82 glycoproteins. Proteomic profiling revealed 27 glycopeptides overexpressed in both NSCLC cell lines, 6 glycopeptides overexpressed only in the EGFR mutant cells and 19 glycopeptides overexpressed only in the KRAS mutant cells. Further investigation of a panel of NSCLC cell lines found that Cellular repressor of E1A-stimulated genes (CREG1) overexpression was closely correlated with KRAS mutation status in NSCLC cells and could be down-regulated by inhibition of KRAS expression. Our results indicate that CREG1 is a down-stream effector of KRAS in a sub-type of NSCLC cells and a novel candidate biomarker or therapeutic target for KRAS mutant NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Glicoproteínas/análise , Proteoma/análise , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Repressoras/metabolismo , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA