Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Food Addit Contam Part B Surveill ; 17(2): 142-152, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38600616

RESUMO

Some synthetic dyes are fraudulently added into spices to appeal visually to consumers. Food regulations in several countries, including the United States, Australia, Japan and the European Union, strictly prohibit the use of unauthorised synthetic dyes in food. Nevertheless, illegal practices persist, where spices contaminated with potentially carcinogenic dyes have been documented, posing potential health risks to consumers. In the present study, 14 synthetic dyes were investigated through liquid chromatography/tandem mass spectrometry in 252 commercially available spices in the Singapore market. In 18 out of these (7.1%) at least 1 illegal dye was detected at concentrations ranging from 0.010 to 114 mg/kg. Besides potential health risks, presence of these adulterants also reflects the economic motivations behind their fraudulent use. Findings in the present study further emphasise the need for increased public awareness, stricter enforcement, and continuous monitoring of illegal synthetic dyes in spices to ensure Singapore's food safety.


Assuntos
Contaminação de Alimentos , Especiarias , Espectrometria de Massas em Tandem , Especiarias/análise , Singapura , Contaminação de Alimentos/análise , Humanos , Corantes/análise , Corantes/química , Corantes de Alimentos/análise
2.
J Proteome Res ; 21(8): 1948-1960, 2022 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-35838755

RESUMO

Englerin A (EA) is a small-molecule natural product with selective cytotoxicity against renal cancer cells. EA has been shown to induce apoptosis and cell death through cell-cycle arrest and/or insulin signaling pathways. However, its biological mode of action or targets in renal cancer remains enigmatic. In this study, we employed advanced mass spectrometry-based phosphoproteomics approaches to identify EA's functional roles in renal cancer. We identified 10,940 phosphorylation sites, of which 706 sites exhibited EA-dependent phosphorylation changes. Integrated analysis of motifs and interaction networks suggested activation of stress-activated kinases including p38 upon EA treatment. Of note, a downstream target of p38, Hsp27, was found to be hyperphosphorylated on multiple sites upon EA treatment. Among these, a novel site Ser65 on Hsp27, which was further validated by targeted proteomics, was shown to be crucial for EA-induced cytotoxicity in renal cancer cells. Taken together, these data reveal the complex signaling cascade that is induced upon EA treatment and importantly provide insights into its effects on downstream molecular signaling.


Assuntos
Proteínas de Choque Térmico HSP27 , Neoplasias Renais , Apoptose , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico HSP27/farmacologia , Humanos , Neoplasias Renais/tratamento farmacológico , Fosforilação , Sesquiterpenos de Guaiano/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/farmacologia
3.
Elife ; 102021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34870592

RESUMO

The Src tyrosine kinase controls cancer-critical protein glycosylation through Golgi to ER relocation of GALNTs enzymes. How Src induces this trafficking event is unknown. Golgi to ER transport depends on the GTP exchange factor (GEF) GBF1 and small GTPase Arf1. Here, we show that Src induces the formation of tubular transport carriers containing GALNTs. The kinase phosphorylates GBF1 on 10 tyrosine residues; two of them, Y876 and Y898, are located near the C-terminus of the Sec7 GEF domain. Their phosphorylation promotes GBF1 binding to the GTPase; molecular modeling suggests partial melting of the Sec7 domain and intramolecular rearrangement. GBF1 mutants defective for these rearrangements prevent binding, carrier formation, and GALNTs relocation, while phosphomimetic GBF1 mutants induce tubules. In sum, Src promotes GALNTs relocation by promoting GBF1 binding to Arf1. Based on residue conservation, similar regulation of GEF-Arf complexes by tyrosine phosphorylation could be a conserved and widespread mechanism.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/genética , Quinases da Família src/genética , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Fosforilação , Transporte Proteico , Quinases da Família src/metabolismo
4.
Nat Commun ; 12(1): 5315, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34493720

RESUMO

Human PAK4 is an ubiquitously expressed p21-activated kinase which acts downstream of Cdc42. Since PAK4 is enriched in cell-cell junctions, we probed the local protein environment around the kinase with a view to understanding its location and substrates. We report that U2OS cells expressing PAK4-BirA-GFP identify a subset of 27 PAK4-proximal proteins that are primarily cell-cell junction components. Afadin/AF6 showed the highest relative biotin labelling and links to the nectin family of homophilic junctional proteins. Reciprocally >50% of the PAK4-proximal proteins were identified by Afadin BioID. Co-precipitation experiments failed to identify junctional proteins, emphasizing the advantage of the BioID method. Mechanistically PAK4 depended on Afadin for its junctional localization, which is similar to the situation in Drosophila. A highly ranked PAK4-proximal protein LZTS2 was immuno-localized with Afadin at cell-cell junctions. Though PAK4 and Cdc42 are junctional, BioID analysis did not yield conventional cadherins, indicating their spatial segregation. To identify cellular PAK4 substrates we then assessed rapid changes (12') in phospho-proteome after treatment with two PAK inhibitors. Among the PAK4-proximal junctional proteins seventeen PAK4 sites were identified. We anticipate mammalian group II PAKs are selective for the Afadin/nectin sub-compartment, with a demonstrably distinct localization from tight and cadherin junctions.


Assuntos
Junções Intercelulares/metabolismo , Proteínas dos Microfilamentos/genética , Nectinas/genética , Proteômica/métodos , Proteína cdc42 de Ligação ao GTP/genética , Quinases Ativadas por p21/genética , Biotina/química , Carbono-Nitrogênio Ligases/genética , Carbono-Nitrogênio Ligases/metabolismo , Linhagem Celular Tumoral , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Junções Intercelulares/genética , Junções Intercelulares/ultraestrutura , Marcação por Isótopo , Espectrometria de Massas , Proteínas dos Microfilamentos/metabolismo , Nectinas/metabolismo , Osteoblastos/metabolismo , Osteoblastos/ultraestrutura , Ligação Proteica , Mapeamento de Interação de Proteínas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteína cdc42 de Ligação ao GTP/metabolismo , Quinases Ativadas por p21/metabolismo
5.
Oncogene ; 38(9): 1508-1519, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30305722

RESUMO

Aberrant activation of Wnt/ß-catenin signaling pathway is essential for the development of AML; however, the mechanistic basis for this dysregulation is unclear. PRL-3 is an oncogenic phosphatase implicated in the development of LSCs. Here, we identified Leo1 as a direct and specific substrate of PRL-3. Serine-dephosphorylated form of Leo1 binds directly to ß-catenin, promoting the nuclear accumulation of ß-catenin and transactivation of TCF/LEF downstream target genes such as cyclin D1 and c-myc. Importantly, overexpression of PRL-3 in AML cells displayed enhanced sensitivity towards ß-catenin inhibition in vitro and in vivo, suggesting that these cells are addicted to ß-catenin signaling. Altogether, our study revealed a novel regulatory role of PRL-3 in the sustenance of aberrant ß-catenin signaling in AML. PRL-3 may serve as a biomarker to select for the subset of AML patients who are likely to benefit from treatment with ß-catenin inhibitors. Our study presents a new avenue of cancer inhibition driven by PRL-3 overexpression or ß-catenin hyperactivation.


Assuntos
Leucemia Mieloide Aguda/genética , Proteínas de Neoplasias/genética , Proteínas Tirosina Fosfatases/genética , Fatores de Transcrição/genética , beta Catenina/genética , Animais , Ciclina D1/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Ligação Proteica/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Wnt/genética , Via de Sinalização Wnt/genética , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cell Signal ; 43: 21-31, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29196224

RESUMO

Activating mutations in the kinase domain of epidermal growth factor receptor (EGFR) leads to the constitutively active kinase, improves the EGFR stability and promotes malignant transformation in lung adenocarcinoma. Despite the clinical significance, the mechanism by which the increased kinase activity stabilizes the receptor is not completely understood. Using SILAC phosphoproteomic approach, we identify that Mig6 is highly phosphorylated at S256 in EGFR mutants (19del and L858R). Loss of Mig6 contributes to the efficient degradation of EGFR wildtype and mutants in lung cancer cells. Mig6 regulates the recruitment of c-Cbl to EGFR as the ablation of Mig6 enables efficient ubiquitination of the EGFR mutants through elevated recruitment of c-Cbl. We show that the cells with activating mutants of EGFR inactivate Mig6 through phosphorylation at S256. Inactivated Mig6 causes inefficient ubiquitination of EGFR, leading to defective degradation of the receptor thus contributing to the increased stability of the receptor. Taken together, we show a novel function of Mig6 in regulating the ubiquitination of EGFR.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenocarcinoma de Pulmão/metabolismo , Proteínas Mutantes/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitinação , Adenocarcinoma de Pulmão/patologia , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Modelos Biológicos , Mutação/genética , Peptídeos/metabolismo , Fosforilação , Fosfosserina/metabolismo , Ligação Proteica , Estabilidade Proteica , Reprodutibilidade dos Testes
7.
Breast Cancer Res ; 19(1): 132, 2017 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-29233185

RESUMO

BACKGROUND: Annexin-1 (ANXA1) plays pivotal roles in regulating various physiological processes including inflammation, proliferation and apoptosis, and deregulation of ANXA1 functions has been associated with tumorigenesis and metastasis events in several types of cancer. Though ANXA1 levels correlate with breast cancer disease status and outcome, its distinct functional involvement in breast cancer initiation and progression remains unclear. We hypothesized that ANXA1-responsive kinase signaling alteration and associated phosphorylation signaling underlie early events in breast cancer initiation events and hence profiled ANXA1-dependent phosphorylation changes in mammary gland epithelial cells. METHODS: Quantitative phosphoproteomics analysis of mammary gland epithelial cells derived from ANXA1-heterozygous and ANXA1-deficient mice was carried out using stable isotope labeling with amino acids in cell culture (SILAC)-based mass spectrometry. Kinase and signaling changes underlying ANXA1 perturbations were derived by upstream kinase prediction and integrated network analysis of altered proteins and phosphoproteins. RESULTS: We identified a total of 8110 unique phosphorylation sites, of which 582 phosphorylation sites on 372 proteins had ANXA1-responsive changes. A majority of these phosphorylation changes occurred on proteins associated with cytoskeletal reorganization spanning the focal adhesion, stress fibers, and also the microtubule network proposing new roles for ANXA1 in regulating microtubule dynamics. Comparative analysis of regulated global proteome and phosphoproteome highlighted key differences in translational and post-translational effects of ANXA1, and suggested closely coordinated rewiring of the cell adhesion network. Kinase prediction analysis suggested activity modulation of calmodulin-dependent protein kinase II (CAMK2), P21-activated kinase (PAK), extracellular signal-regulated kinase (ERK), and IκB kinase (IKK) upon loss of ANXA1. Integrative analysis revealed regulation of the WNT and Hippo signaling pathways in ANXA1-deficient mammary epithelial cells, wherein there is downregulation of transcriptional effects of TEA domain family (TEAD) suggestive of ANXA1-responsive transcriptional rewiring. CONCLUSIONS: The phosphoproteome landscape uncovered several novel perspectives for ANXA1 in mammary gland biology and highlighted its involvement in key signaling pathways modulating cell adhesion and migration that could contribute to breast cancer initiation.


Assuntos
Anexina A1/deficiência , Anexina A1/genética , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Fosfoproteínas/metabolismo , Proteoma , Proteômica , Animais , Adesão Celular , Análise por Conglomerados , Biologia Computacional/métodos , Feminino , Técnicas de Inativação de Genes , Humanos , Camundongos , Mapeamento de Interação de Proteínas , Mapas de Interação de Proteínas , Proteômica/métodos
8.
J Biol Chem ; 292(19): 8092-8100, 2017 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-28280241

RESUMO

Pathogenic Yersinia bacteria cause a range of human diseases. To modulate and evade host immune systems, these yersiniae inject effector proteins into host macrophages. One such protein, the serine/threonine kinase YopO (YpkA in Yersinia pestis), uses monomeric actin as bait to recruit and phosphorylate host actin polymerization-regulating proteins, including the actin-severing protein gelsolin, to disrupt actin filaments and thus impair phagocytosis. However, the YopO phosphorylation sites on gelsolin and the consequences of YopO-mediated phosphorylation on actin remodeling have yet to be established. Here we determined the effects of YopO-mediated phosphorylation on gelsolin and identified its phosphorylation sites by mass spectrometry. YopO phosphorylated gelsolin in the linker region between gelsolin homology domains G3 and G4, which, in the absence of calcium, are compacted but adopt an open conformation in the presence of calcium, enabling actin binding and severing. Using phosphomimetic and phosphodeletion gelsolin mutants, we found that YopO-mediated phosphorylation partially mimics calcium-dependent activation of gelsolin, potentially contributing to a reduction in filamentous actin and altered actin dynamics in phagocytic cells. In summary, this work represents the first report of the functional outcome of serine/threonine phosphorylation in gelsolin regulation and provides critical insight into how YopO disrupts normal gelsolin function to alter host actin dynamics and thus cripple phagocytosis.


Assuntos
Actinas/química , Proteínas de Bactérias/metabolismo , Cálcio/química , Gelsolina/química , Proteínas Serina-Treonina Quinases/metabolismo , Yersinia/metabolismo , Citoesqueleto de Actina/metabolismo , Sítios de Ligação , Humanos , Macrófagos/microbiologia , Espectrometria de Massas , Simulação de Dinâmica Molecular , Mutação , Fagocitose , Fosforilação , Domínios Proteicos , Pirenos/química , Serina/química , Treonina/química
9.
Sci Rep ; 7: 39998, 2017 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-28051168

RESUMO

Yersinia bacteria cause a range of human diseases, including yersiniosis, Far East scarlet-like fever and the plague. Yersiniae modulate and evade host immune defences through injection of Yersinia outer proteins (Yops) into phagocytic cells. One of the Yops, YopO (also known as YpkA) obstructs phagocytosis through disrupting actin filament regulation processes - inhibiting polymerization-promoting signaling through sequestration of Rac/Rho family GTPases and by using monomeric actin as bait to recruit and phosphorylate host actin-regulating proteins. Here we set out to identify mechanisms of specificity in protein phosphorylation by YopO that would clarify its effects on cytoskeleton disruption. We report the MgADP structure of Yersinia enterocolitica YopO in complex with actin, which reveals its active site architecture. Using a proteome-wide kinase-interacting substrate screening (KISS) method, we identified that YopO phosphorylates a wide range of actin-modulating proteins and located their phosphorylation sites by mass spectrometry. Using artificial substrates we clarified YopO's substrate length requirements and its phosphorylation consensus sequence. These findings provide fresh insight into the mechanism of the YopO kinase and demonstrate that YopO executes a specific strategy targeting actin-modulating proteins, across multiple functionalities, to compete for control of their native phospho-signaling, thus hampering the cytoskeletal processes required for macrophage phagocytosis.


Assuntos
Actinas/química , Difosfato de Adenosina/química , Proteínas de Bactérias/química , Proteínas Serina-Treonina Quinases/química , Yersinia enterocolitica , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Especificidade por Substrato
10.
Sci Rep ; 6: 25844, 2016 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-27180971

RESUMO

Despite efforts in the last decade, signaling aberrations associated with obesity remain poorly understood. To dissect molecular mechanisms that define this complex metabolic disorder, we carried out global phosphoproteomic analysis of white adipose tissue (WAT) from mice fed on low-fat diet (LFD) and high-fat diet (HFD). We quantified phosphorylation levels on 7696 peptides, and found significant differential phosphorylation levels in 282 phosphosites from 191 proteins, including various insulin-responsive proteins and metabolic enzymes involved in lipid homeostasis in response to high-fat feeding. Kinase-substrate prediction and integrated network analysis of the altered phosphoproteins revealed underlying signaling modulations during HFD-induced obesity, and suggested deregulation of lipogenic and lipolytic pathways. Mutation of the differentially-regulated novel phosphosite on cytoplasmic acetyl-coA forming enzyme ACSS2 (S263A) upon HFD-induced obesity led to accumulation of serum triglycerides and reduced insulin-responsive AKT phosphorylation as compared to wild type ACSS2, thus highlighting its role in obesity. Altogether, our study presents a comprehensive map of adipose tissue phosphoproteome in obesity and reveals many previously unknown candidate phosphorylation sites for future functional investigation.


Assuntos
Tecido Adiposo Branco/metabolismo , Dieta Hiperlipídica/efeitos adversos , Obesidade/induzido quimicamente , Obesidade/metabolismo , Fosfoproteínas/metabolismo , Células 3T3-L1 , Acetato-CoA Ligase/genética , Acetato-CoA Ligase/metabolismo , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes , Camundongos , Mutação , Peptídeos/química , Fosfoproteínas/química , Mapas de Interação de Proteínas , Proteômica/métodos , Transdução de Sinais , Triglicerídeos/metabolismo
11.
Nat Commun ; 7: 10968, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-27005833

RESUMO

CCAAT/enhancer-binding protein alpha (C/EBPα) is an essential transcription factor for myeloid lineage commitment. Here we demonstrate that acetylation of C/EBPα at lysine residues K298 and K302, mediated at least in part by general control non-derepressible 5 (GCN5), impairs C/EBPα DNA-binding ability and modulates C/EBPα transcriptional activity. Acetylated C/EBPα is enriched in human myeloid leukaemia cell lines and acute myeloid leukaemia (AML) samples, and downregulated upon granulocyte-colony stimulating factor (G-CSF)- mediated granulocytic differentiation of 32Dcl3 cells. C/EBPα mutants that mimic acetylation failed to induce granulocytic differentiation in C/EBPα-dependent assays, in both cell lines and in primary hematopoietic cells. Our data uncover GCN5 as a negative regulator of C/EBPα and demonstrate the importance of C/EBPα acetylation in myeloid differentiation.


Assuntos
Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Regulação Neoplásica da Expressão Gênica , Granulócitos/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide/genética , Mielopoese/genética , Fatores de Transcrição de p300-CBP/genética , Acetilação , Diferenciação Celular/genética , Linhagem Celular Tumoral , Cromatografia Líquida , Ensaio de Desvio de Mobilidade Eletroforética , Fator Estimulador de Colônias de Granulócitos , Granulócitos/citologia , Células HEK293 , Humanos , Immunoblotting , Imunoprecipitação , Leucemia Mieloide/metabolismo , Leucemia Mieloide Aguda/metabolismo , Espectrometria de Massas , Fatores de Transcrição de p300-CBP/metabolismo
12.
Cell Stem Cell ; 17(4): 435-47, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26387754

RESUMO

ELABELA (ELA) is a peptide hormone required for heart development that signals via the Apelin Receptor (APLNR, APJ). ELA is also abundantly secreted by human embryonic stem cells (hESCs), which do not express APLNR. Here we show that ELA signals in a paracrine fashion in hESCs to maintain self-renewal. ELA inhibition by CRISPR/Cas9-mediated deletion, shRNA, or neutralizing antibodies causes reduced hESC growth, cell death, and loss of pluripotency. Global phosphoproteomic and transcriptomic analyses of ELA-pulsed hESCs show that it activates PI3K/AKT/mTORC1 signaling required for cell survival. ELA promotes hESC cell-cycle progression and protein translation and blocks stress-induced apoptosis. INSULIN and ELA have partially overlapping functions in hESC medium, but only ELA can potentiate the TGFß pathway to prime hESCs toward the endoderm lineage. We propose that ELA, acting through an alternate cell-surface receptor, is an endogenous secreted growth factor in human embryos and hESCs that promotes growth and pluripotency.


Assuntos
Células-Tronco Embrionárias Humanas/metabolismo , Hormônios Peptídicos/metabolismo , Transdução de Sinais , Anticorpos Neutralizantes , Receptores de Apelina , Diferenciação Celular , Linhagem Celular , Autorrenovação Celular , Endoderma/citologia , Endoderma/metabolismo , Perfilação da Expressão Gênica , Células-Tronco Embrionárias Humanas/citologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Comunicação Parácrina , Fosfatidilinositol 3-Quinases/metabolismo , Proteômica , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno , Receptores Acoplados a Proteínas G/metabolismo
13.
Glycobiology ; 25(12): 1491-504, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26306634

RESUMO

Transforming growth factor-ß1 (TGF-ß1, Uniprot: P01137) is a heparin-binding protein that has been implicated in a number of physiological processes, including the initiation of chondrogenesis by human mesenchymal stem cells (hMSCs). Here, we identify the molecular features in the protein and in heparin required for binding and their effects on the potentiation of TGF-ß1's activity on hMSCs. Using a proteomics "Protect and Label" approach, lysines K291, K304, K309, K315, K338, K373, K375 and K388 were identified as being directly involved in binding heparin (Data are available via ProteomeXchange with identifier PXD002772). Competition assays in an optical biosensor demonstrated that TGF-ß1 does require N- and 6-O-sulfate groups for binding but that 2-O-sulfate groups are unlikely to underpin the interaction. Heparin-derived oligosaccharides as short as degree of polymerization (dp) 4 have a weak ability to compete for TGF-ß1 binding to heparin, which increases with the length of the oligosaccharide to reach a maximum between dp18 and dp24. In cell-based assays, heparin, 2-O-, 6-O- and N-desulfated re-N-acetylated heparin and oligosaccharides 14-24 saccharides (dp14-24) in length all increased the phosphorylation of mothers against decapentaplegic homolog 2 (SMAD2) after 6 h of stimulation with TGF-ß1. The results provide the structural basis for a model of heparin/heparan sulfate binding to TGF-ß1 and demonstrate that the features in the polysaccharide required for binding are not identical to those required for sustaining the signaling by TGF-ß1 in hMSCs.


Assuntos
Heparina/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/química , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Células Cultivadas , Heparina/química , Humanos , Células-Tronco Mesenquimais/metabolismo , Dados de Sequência Molecular , Ligação Proteica , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
14.
Cell Cycle ; 13(14): 2200-10, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24844324

RESUMO

Degradation of p53 is a cornerstone in the control of its functions as a tumor suppressor. This process is attributed to ubiquitin-dependent modification of p53. In addition to polyubiquitination, we found that p53 is targeted for degradation through ISGylation. Isg15, a ubiquitin-like protein, covalently modifies p53 at 2 sites in the N and C terminus, and ISGylated p53 can be degraded by the 20S proteasome. ISGylation primarily targets a misfolded, dominant-negative p53, and Isg15 deletion in normal cells results in suppression of p53 activity and functions. We propose that Isg15-dependent degradation of p53 represents an alternative mechanism of controlling p53 protein levels, and, thus, it is an attractive pathway for drug discovery.


Assuntos
Citocinas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ubiquitinas/metabolismo , Animais , Citocinas/deficiência , Citocinas/genética , Células HCT116 , Células HEK293 , Humanos , Células MCF-7 , Camundongos Knockout , Complexo de Endopeptidases do Proteassoma/metabolismo , Estabilidade Proteica , Proteólise , Interferência de RNA , Transfecção , Proteína Supressora de Tumor p53/genética , Ubiquitinação , Ubiquitinas/deficiência , Ubiquitinas/genética
15.
Genes Dev ; 27(4): 441-9, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23431056

RESUMO

Endoplasmic reticulum (ER) stress is emerging as a potential contributor to the onset of type 2 diabetes by making cells insulin-resistant. However, our understanding of the mechanisms by which ER stress affects insulin response remains fragmentary. Here we present evidence that the ER stress pathway acts via a conserved signaling mechanism involving the protein kinase PERK to modulate cellular insulin responsiveness. Insulin signaling via AKT reduces activity of FOXO transcription factors. In some cells, PERK can promote insulin responsiveness. However, we found that PERK also acts oppositely via phosphorylation of FOXO to promote FOXO activity. Inhibition of PERK improves cellular insulin responsiveness at the level of FOXO activity. We suggest that the protein kinase PERK may be a promising pharmacological target for ameliorating insulin resistance.


Assuntos
Estresse do Retículo Endoplasmático/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Resistência à Insulina/fisiologia , eIF-2 Quinase/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Células Hep G2 , Humanos , Células MCF-7 , Proteína Oncogênica v-akt/metabolismo , Fosforilação , Transporte Proteico
16.
Chem Commun (Camb) ; (40): 4233-5, 2006 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-17031442

RESUMO

The combined use of advanced mass spectrometry experiments, condensed-phase synthesis of serine and homoserine nitrate ester radical precursors, and high-level ab initio calculations provides a powerful way of examining the fundamental reactivity of radicals derived from peptides.


Assuntos
Aminoácidos/química , Radicais Livres/química , Gases/química , Peptídeos/química , Transporte de Elétrons , Ésteres , Homosserina/química , Serina/química , Espectrometria de Massas por Ionização por Electrospray
17.
Rapid Commun Mass Spectrom ; 19(13): 1797-805, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15945020

RESUMO

Electrospray ionization (ESI) tandem mass spectrometry (MS/MS) of ternary transition metal complexes of [M(L(3))(N)](2+) (where M = copper(II) or platinum(II); L(3) = diethylenetriamine (dien) or 2,2':6',2''-terpyridine (tpy); N = the nucleobases: adenine, guanine, thymine and cytosine; the nucleosides: 2'deoxyadenosine, 2'deoxyguanosine, 2'deoxythymine, 2'deoxycytidine; the nucleotides: 2'deoxyadenosine 5'-monophosphate, 2'deoxyguanosine 5'-monophosphate, 2'deoxythymine 5'-monophosphate, 2'deoxycytidine 5'-monophosphate) was examined as a means of forming radical cations of the constituents of nucleic acids in the gas phase. In general, sufficient quantities of the ternary complexes [M(L(3))(N)](2+) could be formed for MS/MS studies by subjecting methanolic solutions of mixtures of a metal salt [M(L(3))X(2)] (where M = Cu(II) or Pt(II); L(3) = dien or tpy; X = Cl or NO(3)) and N to ESI. The only exceptions were thymine and its derivatives, which failed to form sufficient abundances of [M(L(3))(N)](2+) ions when: (a) M = Pt(II) and L(3) = dien or tpy; (b) M = Cu(II) and L(3) = dien. In some instances higher oligomeric complexes were formed; e.g., [Pt(tpy)(dG)(n)](2+) (n = 1-13). Each of the ternary complexes [M(L(3))(N)](2+) was mass-selected and then subjected to collision-induced dissociation (CID) in a quadrupole ion trap. The types of fragmentation reactions observed for these complexes depend on the nature of all three components (metal, auxiliary ligand and nucleic acid constituent) and can be classified into: (i) a redox reaction which results in the formation of the radical cation of the nucleic acid constituent, N(+.); (ii) loss of the nucleic acid constituent in its protonated form; and (iii) fragmentation of the nucleic acid constituent. Only the copper complexes yielded radical cations of the nucleic acid constituent, with [Cu(tpy)(N)](2+) being the preferred complex due to suppression, in this case, of the loss of the nucleobase in its protonated form. The yields of the radical cations of the nucleobases from the copper complexes follow the order of their ionization potentials (IPs): G (lowest IP) > A > C > T (highest IP). Sufficient yields of the radical cations of each of the nucleobases allowed their CID reactions (in MS(3) experiments) to be compared to their even-electron counterparts.


Assuntos
Cátions/química , Metais/química , Ácidos Nucleicos/química , Radicais Livres/química , Indicadores e Reagentes , Ligantes , Solventes , Espectrometria de Massas por Ionização por Electrospray
18.
Dalton Trans ; (20): 3199-204, 2004 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-15483701

RESUMO

A series of ternary copper(II) complexes of the type [Cu(II)(L)(M)](2+), where M represents the hexapeptides GGGFLR, YGGFLR and WGGFLR and L a set of 12 nitrogen donor ligands have been evaluated for their ability to form cationic peptide radicals, M(+)*, in the gas phase. Although the fragmentation chemistry of these ions is complex, two main conclusions emerge: (i) Complexes containing a tri- or tetra-dentate ligand were found to be more effective at producing the peptide radical because in these instances competitive loss of the ligand from the complex is inhibited; (ii) The ligands ought not possess any acidic protons in order to prevent competitive loss of the protonated peptide, [M + H](+). There is significant interaction of the N-terminal aromatic residues in YGGFLR and WGGLFR with the copper(ii) ion in several of the complexes as revealed by the formation of [Cu(I)(L)(p-quinomethide)](+) and [Cu(I)(L)(3-methyleneindoline)](+) fragment ions. Following its dissociation from the ternary complex, CID of the YGGFLR(+)* radical cation shows a dependence on the ligand in the complex from which it was formed. This 'memory effect' most likely reflects differences in the coordinated peptide structure induced by the ligand in the precursor complex which are maintained following dissociation.


Assuntos
Cobre/química , Compostos Organometálicos/química , Peptídeos/química , Ligantes , Espectrometria de Massas , Compostos Organometálicos/síntese química
19.
Rapid Commun Mass Spectrom ; 18(11): 1221-6, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15164352

RESUMO

The source of protons associated with the ligand loss channel of HX((n - 1)+) from [Pt(II)(dien)X](n+) (X = Cl, Br and I for n = 1 and X = NC(5)H(5) for n = 2) in the gas phase was investigated by deuterium-labelling studies. The results of these studies indicate that these protons originate from both the amino groups and the carbon backbone of the dien ligand. In some instances (e.g. X = Br and I), the protons lost from the carbon backbone can be even more abundant than the protons lost from the amino groups. The gas-phase substitution reactions of coordinatively saturated [Pt(II)(L(3))L(a)](2+) complexes (L(3) = tpy or dien) were also examined using ion-molecule reactions. The outcome of the ion-molecule reactions depends on both the ancillary ligand (L(3)) as well as the leaving group (L(a)). [Pt(II)(tpy)L(a)](2+) complexes undergo substitution reactions, with a faster rate when L(a) is a good leaving group, while the [Pt(II)(dien)L(a)](2+) complex undergoes a proton transfer reaction.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA