Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Plant Cell ; 31(9): 2010-2034, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31266899

RESUMEN

The order of enzymatic activity across Golgi cisternae is essential for complex molecule biosynthesis. However, an inability to separate Golgi cisternae has meant that the cisternal distribution of most resident proteins, and their underlying localization mechanisms, are unknown. Here, we exploit differences in surface charge of intact cisternae to perform separation of early to late Golgi subcompartments. We determine protein and glycan abundance profiles across the Golgi; over 390 resident proteins are identified, including 136 new additions, with over 180 cisternal assignments. These assignments provide a means to better understand the functional roles of Golgi proteins and how they operate sequentially. Protein and glycan distributions are validated in vivo using high-resolution microscopy. Results reveal distinct functional compartmentalization among resident Golgi proteins. Analysis of transmembrane proteins shows several sequence-based characteristics relating to pI, hydrophobicity, Ser abundance, and Phe bilayer asymmetry that change across the Golgi. Overall, our results suggest that a continuum of transmembrane features, rather than discrete rules, guide proteins to earlier or later locations within the Golgi stack.


Asunto(s)
Aparato de Golgi/metabolismo , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Aparato de Golgi/ultraestructura , Interacciones Hidrofóbicas e Hidrofílicas , Membranas Intracelulares , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Polisacáridos/química , Polisacáridos/metabolismo , Proteoma
2.
Nucleic Acids Res ; 47(9): 4569-4585, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30838409

RESUMEN

UNG is the major uracil-DNA glycosylase in mammalian cells and is involved in both error-free base excision repair of genomic uracil and mutagenic uracil-processing at the antibody genes. However, the regulation of UNG in these different processes is currently not well understood. The UNG gene encodes two isoforms, UNG1 and UNG2, each possessing unique N-termini that mediate translocation to the mitochondria and the nucleus, respectively. A strict subcellular localization of each isoform has been widely accepted despite a lack of models to study them individually. To determine the roles of each isoform, we generated and characterized several UNG isoform-specific mouse and human cell lines. We identified a distinct UNG1 isoform variant that is targeted to the cell nucleus where it supports antibody class switching and repairs genomic uracil. We propose that the nuclear UNG1 variant, which in contrast to UNG2 lacks a PCNA-binding motif, may be specialized to act on ssDNA through its ability to bind RPA. RPA-coated ssDNA regions include both transcribed antibody genes that are targets for deamination by AID and regions in front of the moving replication forks. Our findings provide new insights into the function of UNG isoforms in adaptive immunity and DNA repair.


Asunto(s)
ADN Glicosilasas/genética , Reparación del ADN/genética , Cambio de Clase de Inmunoglobulina/genética , Recombinación Genética/genética , Uracil-ADN Glicosidasa/genética , Animales , Sistemas CRISPR-Cas/genética , Línea Celular , Núcleo Celular/genética , Replicación del ADN/genética , ADN de Cadena Simple/genética , Técnicas de Inactivación de Genes , Genoma/genética , Humanos , Ratones , Antígeno Nuclear de Célula en Proliferación/genética , Isoformas de Proteínas/genética , Uracilo/metabolismo
3.
J Transl Med ; 18(1): 159, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32264925

RESUMEN

BACKGROUND: HDAC inhibitors (HDACi) belong to a new group of chemotherapeutics that are increasingly used in the treatment of lymphocyte-derived malignancies, but their mechanisms of action remain poorly understood. Here we aimed to identify novel protein targets of HDACi in B- and T-lymphoma cell lines and to verify selected candidates across several mammalian cell lines. METHODS: Jurkat T- and SUDHL5 B-lymphocytes were treated with the HDACi SAHA (vorinostat) prior to SILAC-based quantitative proteome analysis. Selected differentially expressed proteins were verified by targeted mass spectrometry, RT-PCR and western analysis in multiple mammalian cell lines. Genomic uracil was quantified by LC-MS/MS, cell cycle distribution analyzed by flow cytometry and class switch recombination monitored by FACS in murine CH12F3 cells. RESULTS: SAHA treatment resulted in differential expression of 125 and 89 proteins in Jurkat and SUDHL5, respectively, of which 19 were commonly affected. Among these were several oncoproteins and tumor suppressors previously not reported to be affected by HDACi. Several key enzymes determining the cellular dUTP/dTTP ratio were downregulated and in both cell lines we found robust depletion of UNG2, the major glycosylase in genomic uracil sanitation. UNG2 depletion was accompanied by hyperacetylation and mediated by increased proteasomal degradation independent of cell cycle stage. UNG2 degradation appeared to be ubiquitous and was observed across several mammalian cell lines of different origin and with several HDACis. Loss of UNG2 was accompanied by 30-40% increase in genomic uracil in freely cycling HEK cells and reduced immunoglobulin class-switch recombination in murine CH12F3 cells. CONCLUSION: We describe several oncoproteins and tumor suppressors previously not reported to be affected by HDACi in previous transcriptome analyses, underscoring the importance of proteome analysis to identify cellular effectors of HDACi treatment. The apparently ubiquitous depletion of UNG2 and PCLAF establishes DNA base excision repair and translesion synthesis as novel pathways affected by HDACi treatment. Dysregulated genomic uracil homeostasis may aid interpretation of HDACi effects in cancer cells and further advance studies on this class of inhibitors in the treatment of APOBEC-expressing tumors, autoimmune disease and HIV-1.


Asunto(s)
Inhibidores de Histona Desacetilasas , Uracilo , Animales , Línea Celular , Cromatografía Liquida , Genómica , Inhibidores de Histona Desacetilasas/farmacología , Ratones , Proteínas Oncogénicas , Linfocitos T , Espectrometría de Masas en Tándem , Uracilo/farmacología
4.
Hum Mol Genet ; 26(6): 1031-1040, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28007905

RESUMEN

Recessive loss of function of the neuronal ubiquitin hydrolase UCHL1 has been implicated in early-onset progressive neurodegeneration (MIM no. 615491), so far only in one family. In this study a second family is characterized, and the functional consequences of the identified mutations in UCHL1 are explored. Three siblings developed childhood-onset optic atrophy, followed by spasticity and ataxia. Whole exome sequencing identified compound heterozygous variants in UCHL1, c.533G > A (p.Arg178Gln) and c.647C > A (p.Ala216Asp), cosegregating with the phenotype. Enzymatic activity of purified recombinant proteins analysed by ubiquitin hydrolase assays showed a 4-fold increased hydrolytic activity of the recombinant UCHL1 mutant Arg178Gln compared to wild type, whereas the Ala216Asp protein was insoluble. Structural 3D analysis of UCHL1 by computer modelling suggests that Arg178 is a rate-controlling residue in catalysis which is partly abolished in the Arg178Gln mutant and, consequently, the Arg178Gln mutant increases the enzymatic turnover. UCHL1 protein levels in fibroblasts measured by targeted mass spectrometry showed a total amount of UCHL1 in control fibroblasts about 4-fold higher than in the patients. Hence, studies of the identified missense variants reveal surprisingly different functional consequences as the insoluble Ala216Asp variant leads to loss of function, whereas the Arg178Gln leads to increased enzyme activity. The reported patients have remarkably preserved cognition, and we propose that the increased enzyme activity of the Arg178Gln variant offers a protective effect on cognitive function. This study establishes the importance of UCHL1 in neurodegeneration, provides new mechanistic insight about ubiquitin processing, and underlines the complexity of the different roles of UCHL1.


Asunto(s)
Ataxia/genética , Degeneración Nerviosa/genética , Atrofia Óptica/genética , Proteínas Recombinantes/genética , Ubiquitina Tiolesterasa/genética , Anciano , Animales , Ataxia/diagnóstico por imagen , Ataxia/fisiopatología , Modelos Animales de Enfermedad , Exoma , Femenino , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Mutación , Degeneración Nerviosa/diagnóstico por imagen , Degeneración Nerviosa/fisiopatología , Atrofia Óptica/diagnóstico por imagen , Atrofia Óptica/fisiopatología , Conformación Proteica , Proteínas Recombinantes/química , Hermanos , Relación Estructura-Actividad , Ubiquitina Tiolesterasa/química
5.
Nucleic Acids Res ; 45(14): 8291-8301, 2017 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-28575236

RESUMEN

Base lesions in DNA can stall the replication machinery or induce mutations if bypassed. Consequently, lesions must be repaired before replication or in a post-replicative process to maintain genomic stability. Base excision repair (BER) is the main pathway for repair of base lesions and is known to be associated with DNA replication, but how BER is organized during replication is unclear. Here we coupled the iPOND (isolation of proteins on nascent DNA) technique with targeted mass-spectrometry analysis, which enabled us to detect all proteins required for BER on nascent DNA and to monitor their spatiotemporal orchestration at replication forks. We demonstrate that XRCC1 and other BER/single-strand break repair (SSBR) proteins are enriched in replisomes in unstressed cells, supporting a cellular capacity of post-replicative BER/SSBR. Importantly, we identify for the first time the DNA glycosylases MYH, UNG2, MPG, NTH1, NEIL1, 2 and 3 on nascent DNA. Our findings suggest that a broad spectrum of DNA base lesions are recognized and repaired by BER in a post-replicative process.


Asunto(s)
Roturas del ADN de Cadena Simple , Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN , Replicación del ADN , ADN/genética , Línea Celular Tumoral , ADN/metabolismo , ADN Glicosilasas/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Proteínas de Unión al ADN/metabolismo , Desoxirribonucleasa (Dímero de Pirimidina)/metabolismo , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Espectrometría de Masas/métodos , N-Glicosil Hidrolasas/metabolismo , Transducción de Señal/genética , Factores de Tiempo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
7.
Basic Res Cardiol ; 110(4): 44, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26112154

RESUMEN

Diabetes mellitus (DM) increases the risk of heart failure after myocardial infarction (MI), and aggravates ventricular arrhythmias in heart failure patients. Although exercise training improves cardiac function in heart failure, it is still unclear how it benefits the diabetic heart after MI. To study the effects of aerobic interval training on cardiac function, susceptibility to inducible ventricular arrhythmias and cardiomyocyte calcium handling in DM mice after MI (DM-MI). Male type 2 DM mice (C57BLKS/J Lepr (db) /Lepr (db) ) underwent MI or sham surgery. One group of DM-MI mice was submitted to aerobic interval training running sessions during 6 weeks. Cardiac function and structure were assessed by echocardiography and magnetic resonance imaging, respectively. Ventricular arrhythmias were induced by high-frequency cardiac pacing in vivo. Protein expression was measured by Western blot. DM-MI mice displayed increased susceptibility for inducible ventricular arrhythmias and impaired diastolic function when compared to wild type-MI, which was associated with disruption of cardiomyocyte calcium handling and increased calcium leak from the sarcoplasmic reticulum. High-intensity exercise recovered cardiomyocyte function in vitro, reduced sarcoplasmic reticulum diastolic calcium leak and significantly reduced the incidence of inducible ventricular arrhythmias in vivo in DM-MI mice. Exercise training also normalized the expression profile of key proteins involved in cardiomyocyte calcium handling, suggesting a potential molecular mechanism for the benefits of exercise in DM-MI mice. High-intensity aerobic exercise training recovers cardiomyocyte function and reduces inducible ventricular arrhythmias in infarcted diabetic mice.


Asunto(s)
Arritmias Cardíacas/prevención & control , Diabetes Mellitus Tipo 2/complicaciones , Infarto del Miocardio/complicaciones , Condicionamiento Físico Animal , Animales , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Contracción Miocárdica , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Retículo Sarcoplasmático/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/fisiología , Función Ventricular Izquierda
8.
Biotechnol Bioeng ; 112(1): 111-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24981116

RESUMEN

To aid in debugging efforts to increase yield, titer, and productivity of engineered metabolic pathways, computational models are increasingly needed to predict how changes in experimentally manipulable variables such as enzyme expression map to changes in pathway flux. Here, an ordinary differential equation model is developed for a heterologous mevalonate pathway in E. coli using kinetic parameters culled from literature and enzyme concentrations derived from Selective Reaction Monitoring Mass Spectrometry (SRM-MS). To identify parameters most important to further experimental investigation, a global sensitivity analysis was performed, which pointed to amorphadiene synthase activity as the main limiting factor for amorphadiene production. Furthermore, the model predicted that in this local enzyme expression regime, the overall pathway flux is insensitive to farnesyl pyrophosphate (FPP)-mediated inhibition of mevalonate kinase, not supporting a hypothesis that had previously been posited to be limiting amorphadiene production. To test these predictions experimentally, two strains were constructed: (1) a strain containing a homologous mevalonate kinase with weaker feedback inhibition, and (2) a strain with greater amorphadiene synthase expression. The experimental results validate the qualitative model hypotheses and accurately match the predicted productivities for the two strains, particularly when an in vivo-derived kcat for amorphadiene synthase was substituted for the literature value. These results demonstrate the utility of using kinetic representations of engineered metabolic pathways parameterized with experimentally derived protein concentrations and enzyme kinetic constants to predict productivities and test hypotheses about engineering strategies.


Asunto(s)
Escherichia coli/metabolismo , Ingeniería Metabólica/métodos , Ácido Mevalónico/metabolismo , Proteómica/métodos , Transferasas Alquil y Aril/genética , Transferasas Alquil y Aril/metabolismo , Escherichia coli/genética , Retroalimentación Fisiológica , Cinética , Terpenos/metabolismo
10.
Metab Eng ; 26: 48-56, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25205128

RESUMEN

Transformation of engineered Escherichia coli into a robust microbial factory is contingent on precise control of metabolism. Yet, the throughput of omics technologies used to characterize cell components has lagged far behind our ability to engineer novel strains. To expand the utility of quantitative proteomics for metabolic engineering, we validated and optimized targeted proteomics methods for over 400 proteins from more than 20 major pathways in E. coli metabolism. Complementing these methods, we constructed a series of synthetic genes to produce concatenated peptides (QconCAT) for absolute quantification of the proteins and made them available through the Addgene plasmid repository (www.addgene.org). To facilitate high sample throughput, we developed a fast, analytical-flow chromatography method using a 5.5-min gradient (10 min total run time). Overall this toolkit provides an invaluable resource for metabolic engineering by increasing sample throughput, minimizing development time and providing peptide standards for absolute quantification of E. coli proteins.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Perfilación de la Expresión Génica/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Péptidos/metabolismo , Ingeniería de Proteínas/métodos , Péptidos/genética , Mapeo de Interacción de Proteínas/métodos , Proteómica/métodos
11.
EMBO J ; 27(1): 51-61, 2008 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-18079698

RESUMEN

Human UNG2 is a multifunctional glycosylase that removes uracil near replication forks and in non-replicating DNA, and is important for affinity maturation of antibodies in B cells. How these diverse functions are regulated remains obscure. Here, we report three new phosphoforms of the non-catalytic domain that confer distinct functional properties to UNG2. These are apparently generated by cyclin-dependent kinases through stepwise phosphorylation of S23, T60 and S64 in the cell cycle. Phosphorylation of S23 in late G1/early S confers increased association with replication protein A (RPA) and replicating chromatin and markedly increases the catalytic turnover of UNG2. Conversely, progressive phosphorylation of T60 and S64 throughout S phase mediates reduced binding to RPA and flag UNG2 for breakdown in G2 by forming a cyclin E/c-myc-like phosphodegron. The enhanced catalytic turnover of UNG2 p-S23 likely optimises the protein to excise uracil along with rapidly moving replication forks. Our findings may aid further studies of how UNG2 initiates mutagenic rather than repair processing of activation-induced deaminase-generated uracil at Ig loci in B cells.


Asunto(s)
Ciclo Celular/fisiología , ADN Glicosilasas/metabolismo , Proteína de Replicación A/metabolismo , Secuencia de Aminoácidos , Animales , Catálisis , Bovinos , ADN Glicosilasas/química , ADN Glicosilasas/genética , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Fosfoproteínas/química , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína de Replicación A/fisiología , Serina/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Treonina/metabolismo , Uracilo/metabolismo
12.
Nucleic Acids Res ; 38(19): 6447-55, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20525795

RESUMEN

Human AlkB homologues ABH2 and ABH3 repair 1-methyladenine and 3-methylcytosine in DNA/RNA by oxidative demethylation. The enzymes have similar overall folds and active sites, but are functionally divergent. ABH2 efficiently demethylates both single- and double-stranded (ds) DNA, whereas ABH3 has a strong preference for single-stranded DNA and RNA. We find that divergent F1 ß-hairpins in proximity of the active sites of ABH2 and ABH3 are central for substrate specificities. Swapping F1 hairpins between the enzymes resulted in hybrid proteins resembling the donor proteins. Surprisingly, mutation of the intercalating residue F102 had little effect on activity, while the double mutant V101A/F102A was catalytically impaired. These residues form part of an important hydrophobic network only present in ABH2. In this functionally important network, F124 stacks with the flipped out base while L157 apparently functions as a buffer stop to position the lesion in the catalytic pocket for repair. F1 in ABH3 contains charged and polar residues preventing use of dsDNA substrate. Thus, E123 in ABH3 corresponds to F102 in ABH2 and the E123F-variant gained capacity to repair dsDNA with no loss in single strand repair capacity. In conclusion, divergent sequences outside of the active site determine substrate specificities of ABH2 and ABH3.


Asunto(s)
Enzimas Reparadoras del ADN/química , ADN de Cadena Simple/metabolismo , ADN/metabolismo , Dioxigenasas/química , Dioxigenasa Dependiente de Alfa-Cetoglutarato, Homólogo 2 de AlkB , Dioxigenasa Dependiente de Alfa-Cetoglutarato, Homólogo 3 de AlkB , Dominio Catalítico , ADN/química , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , ADN de Cadena Simple/química , Dioxigenasas/genética , Dioxigenasas/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Estructura Secundaria de Proteína , Especificidad por Sustrato
13.
Photochem Photobiol Sci ; 10(7): 1137-45, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21448498

RESUMEN

Photodynamic therapy (PDT) using blue light and the potent precursor for protoporphyrin IX, hexyl aminolevulinate (HAL), has been shown to induce apoptosis and necrosis in cancer cells, but the mechanism remains obscure. In the present study, we examined protein carbonylation, expression levels and post-translational modifications in rat bladder cells (AY-27) after PDT with HAL. Altered levels of expression and/or post-translational modifications induced by PDT were observed for numerous proteins, including proteins required for cell mobility, energy supply, cell survival and cell death pathways, by using two-dimensional difference gel electrophoresis (2D-DIGE) and mass spectrometry (MS). Moreover, 10 carbonylated proteins associated with cytoskeleton, transport, oxidative stress response, protein biosynthesis and stability, and DNA repair were identified using immunoprecipitation, two-dimensional gel electrophoresis and MS. Overall, the results indicate that HAL-mediated PDT triggers a complex cellular response involving several biological pathways. Our findings may account for the elucidation of mechanisms modulated by PDT, paving the way to improve clinic PDT-efficacy.


Asunto(s)
Ácido Aminolevulínico/análogos & derivados , Apoptosis/efectos de los fármacos , Fármacos Fotosensibilizantes/farmacología , Carbonilación Proteica/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteoma/metabolismo , Ácido Aminolevulínico/farmacología , Animales , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Reparación del ADN , Electroforesis en Gel Bidimensional , Fotoquimioterapia , Carbonilación Proteica/efectos de la radiación , Procesamiento Proteico-Postraduccional/efectos de la radiación , Ratas , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico
14.
Cell Rep ; 30(12): 4165-4178.e7, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32209476

RESUMEN

Oxidation resistance gene 1 (OXR1) protects cells against oxidative stress. We find that male mice with brain-specific isoform A knockout (Oxr1A-/-) develop fatty liver. RNA sequencing of male Oxr1A-/- liver indicates decreased growth hormone (GH) signaling, which is known to affect liver metabolism. Indeed, Gh expression is reduced in male mice Oxr1A-/- pituitary gland and in rat Oxr1A-/- pituitary adenoma cell-line GH3. Oxr1A-/- male mice show reduced fasting-blood GH levels. Pull-down and proximity ligation assays reveal that OXR1A is associated with arginine methyl transferase PRMT5. OXR1A-depleted GH3 cells show reduced symmetrical dimethylation of histone H3 arginine 2 (H3R2me2s), a product of PRMT5 catalyzed methylation, and chromatin immunoprecipitation (ChIP) of H3R2me2s shows reduced Gh promoter enrichment. Finally, we demonstrate with purified proteins that OXR1A stimulates PRMT5/MEP50-catalyzed H3R2me2s. Our data suggest that OXR1A is a coactivator of PRMT5, regulating histone arginine methylation and thereby GH production within the pituitary gland.


Asunto(s)
Arginina/metabolismo , Histonas/metabolismo , Proteínas Mitocondriales/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Animales , Encéfalo/metabolismo , Línea Celular , Hígado Graso/genética , Hígado Graso/patología , Femenino , Regulación de la Expresión Génica , Hormona del Crecimiento/sangre , Hormona del Crecimiento/metabolismo , Hormonas/metabolismo , Inmunidad/genética , Hígado/metabolismo , Hígado/patología , Masculino , Metilación , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/química , Proteínas Mitocondriales/deficiencia , Especificidad de Órganos , Hipófisis/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Dominios Proteicos , Ratas , Receptores de Somatotropina/metabolismo , Factor de Transcripción STAT5/metabolismo , Relación Estructura-Actividad , Transcriptoma/genética
15.
Mol Aspects Med ; 28(3-4): 276-306, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17590428

RESUMEN

Uracil is usually an inappropriate base in DNA, but it is also a normal intermediate during somatic hypermutation (SHM) and class switch recombination (CSR) in adaptive immunity. In addition, uracil is introduced into retroviral DNA by the host as part of a defence mechanism. The sources of uracil in DNA are spontaneous or enzymatic deamination of cytosine (U:G mispairs) and incorporation of dUTP (U:A pairs). Uracil in DNA is removed by a uracil-DNA glycosylase. The major ones are nuclear UNG2 and mitochondrial UNG1 encoded by the UNG-gene, and SMUG1 that also removes oxidized pyrimidines, e.g. 5-hydroxymethyluracil. The other ones are TDG that removes U and T from mismatches, and MBD4 that removes U from CpG contexts. UNG2 is found in replication foci during the S-phase and has a distinct role in repair of U:A pairs, but it is also important in U:G repair, a function shared with SMUG1. SHM is initiated by activation-induced cytosine deaminase (AID), followed by removal of U by UNG2. Humans lacking UNG2 suffer from recurrent infections and lymphoid hyperplasia, and have skewed SHM and defective CSR, resulting in elevated IgM and strongly reduced IgG, IgA and IgE. UNG-defective mice also develop B-cell lymphoma late in life. In the defence against retrovirus, e.g. HIV-1, high concentrations of dUTP in the target cells promotes misincorporation of dUMP-, and host cell APOBEC proteins may promote deamination of cytosine in the viral DNA. This facilitates degradation of viral DNA by UNG2 and AP-endonuclease. However, viral proteins Vif and Vpr counteract this defense by mechanisms that are now being revealed. In conclusion, uracil in DNA is both a mutagenic burden and a tool to modify DNA for diversity or degradation.


Asunto(s)
Disparidad de Par Base , ADN/química , Uracilo/química , Animales , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Citosina/química , Reparación del ADN , Replicación del ADN , Humanos , Estructura Molecular , Virus/genética
16.
PLoS One ; 12(9): e0184514, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28902909

RESUMEN

The cytoprotective protein clusterin is often dysregulated during tumorigenesis, and in the stomach, upregulation of clusterin marks emergence of the oxyntic atrophy (loss of acid-producing parietal cells)-associated spasmolytic polypeptide-expressing metaplasia (SPEM). The hormone gastrin is important for normal function and maturation of the gastric oxyntic mucosa and hypergastrinemia might be involved in gastric carcinogenesis. Gastrin induces expression of clusterin in adenocarcinoma cells. In the present study, we examined the expression patterns and gastrin-mediated regulation of clusterin in gastric tissue from: humans; rats treated with proton pump (H+/K+-ATPase) inhibitors and/or a gastrin receptor (CCK2R) antagonist; H+/K+-ATPase ß-subunit knockout (H/K-ß KO) mice; and Mongolian gerbils infected with Helicobacter pylori and given a CCK2R antagonist. Biological function of secretory clusterin was studied in human gastric cancer cells. Clusterin was highly expressed in neuroendocrine cells in normal oxyntic mucosa of humans and rodents. In response to hypergastrinemia, expression of clusterin increased significantly and its localization shifted to basal groups of proliferative cells in the mucous neck cell-chief cell lineage in all animal models. That shift was partially inhibited by antagonizing the CCK2R in rats and gerbils. The oxyntic mucosa of H/K-ß KO mice contained areas with clusterin-positive mucous cells resembling SPEM. In gastric adenocarcinomas, clusterin mRNA expression was higher in diffuse tumors containing signet ring cells compared with diffuse tumors without signet ring cells, and clusterin seemed to be secreted by tumor cells. In gastric cancer cell lines, gastrin increased secretion of clusterin, and both gastrin and secretory clusterin promoted survival after starvation- and chemotherapy-induced stress. Overall, our results indicate that clusterin is overexpressed in hypergastrinemic rodent models of oxyntic preneoplasia and stimulates gastric cancer cell survival.


Asunto(s)
Clusterina/fisiología , Regulación Neoplásica de la Expresión Génica , Células Parietales Gástricas/patología , Neoplasias Gástricas/patología , Anciano , Anciano de 80 o más Años , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular Tumoral , Clusterina/genética , Clusterina/metabolismo , Femenino , Gastrinas/metabolismo , Gastrinas/fisiología , Perfilación de la Expresión Génica , Gerbillinae , Humanos , Masculino , Ratones Noqueados , Persona de Mediana Edad , Células Parietales Gástricas/metabolismo , Inhibidores de la Bomba de Protones/farmacología , Ratas , Ratas Sprague-Dawley , Receptor de Colecistoquinina B/antagonistas & inhibidores , Neoplasias Gástricas/metabolismo
17.
Oncotarget ; 8(12): 19427-19442, 2017 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-28038447

RESUMEN

Plasma cell leukemia is a rare and aggressive plasma cell neoplasm that may either originate de novo (primary PCL) or by leukemic transformation of multiple myeloma (MM) to secondary PCL (sPCL). The prognosis of sPCL is very poor, and currently no standard treatment is available due to lack of prospective clinical studies. In an attempt to elucidate factors contributing to transformation, we have performed super-SILAC quantitative proteome profiling of malignant plasma cells collected from the same patient at both the MM and sPCL stages of the disease. 795 proteins were found to be differentially expressed in the MM and sPCL samples. Gene ontology analysis indicated a metabolic shift towards aerobic glycolysis in sPCL as well as marked down-regulation of enzymes involved in glycan synthesis, potentially mediating altered glycosylation of surface receptors. There was no significant change in overall genomic 5-methylcytosine or 5-hydroxymethylcytosine at the two stages, indicating that epigenetic dysregulation was not a major driver of transformation to sPCL. The present study constitutes the first attempt to provide a comprehensive map of the altered protein expression profile accompanying transformation of MM to sPCL in a single patient, identifying several candidate proteins that can be targeted by currently available small molecule drugs. Our dataset furthermore constitutes a reference dataset for further proteomic analysis of sPCL transformation.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Transformación Celular Neoplásica/patología , Regulación Leucémica de la Expresión Génica , Leucemia de Células Plasmáticas/patología , Mieloma Múltiple/patología , Proteoma/análisis , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Metilación de ADN , Perfilación de la Expresión Génica , Humanos , Leucemia de Células Plasmáticas/genética , Leucemia de Células Plasmáticas/metabolismo , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Proteómica/métodos , Células Tumorales Cultivadas
18.
DNA Repair (Amst) ; 25: 60-71, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25486549

RESUMEN

The most common mutations in cancer are C to T transitions, but their origin has remained elusive. Recently, mutational signatures of APOBEC-family cytosine deaminases were identified in many common cancers, suggesting off-target deamination of cytosine to uracil as a common mutagenic mechanism. Here we present evidence from mass spectrometric quantitation of deoxyuridine in DNA that shows significantly higher genomic uracil content in B-cell lymphoma cell lines compared to non-lymphoma cancer cell lines and normal circulating lymphocytes. The genomic uracil levels were highly correlated with AID mRNA and protein expression, but not with expression of other APOBECs. Accordingly, AID knockdown significantly reduced genomic uracil content. B-cells stimulated to express endogenous AID and undergo class switch recombination displayed a several-fold increase in total genomic uracil, indicating that B cells may undergo widespread cytosine deamination after stimulation. In line with this, we found that clustered mutations (kataegis) in lymphoma and chronic lymphocytic leukemia predominantly carry AID-hotspot mutational signatures. Moreover, we observed an inverse correlation of genomic uracil with uracil excision activity and expression of the uracil-DNA glycosylases UNG and SMUG1. In conclusion, AID-induced mutagenic U:G mismatches in DNA may be a fundamental and common cause of mutations in B-cell malignancies.


Asunto(s)
Citidina Desaminasa/metabolismo , ADN de Neoplasias/metabolismo , Linfoma de Células B/genética , Mutación , Uracilo/metabolismo , Disparidad de Par Base , Línea Celular Tumoral , Citosina/metabolismo , Reparación del ADN , Desaminación , Técnicas de Silenciamiento del Gen , Humanos , Cambio de Clase de Inmunoglobulina , Linfoma de Células B/enzimología , Linfoma de Células B/metabolismo , Mutación Puntual , Uracil-ADN Glicosidasa/metabolismo
19.
Obes Surg ; 23(1): 39-49, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22949011

RESUMEN

BACKGROUND: Ileal interposition-sleeve gastrectomy (II-SG) has been developed as a metabolic surgery based on the hindgut hypothesis. The aim of the present study was to test this hypothesis by studying the eating behavior, metabolic changes, and glucagon-like peptide-1 (GLP-1)-producing cells in rat models. METHODS: Male Sprague-Dawley rats were subjected to laparotomy, II, SG, or II-SG. Eating behavior and metabolic parameters were monitored by an open-circuit indirect calorimeter designed for a comprehensive laboratory animal monitoring system. GLP-1-producing cells were examined by quantitative immunohistochemistry. RESULTS: After II alone, satiety ratio, i.e., intermeal interval/meal size, was reduced, while calorie intake was increased at 2 and 6 weeks postoperatively. Respiratory exchange ratio, VCO(2)/VO(2), was increased to above 1.0 (i.e., carbohydrate metabolism) during both daytime and nighttime at 2 weeks postoperatively. After SG alone, GLP-1-producing cells were increased in the pancreatic islets (in terms of volume density), but not in the ileum (number/mm). After II-SG, the rate of eating was reduced, while meal duration (min) was increased during both daytime and nighttime at 2 weeks postoperatively. GLP-1-producing cells were increased by about 2.5-fold in the interposed ileum and also increased to the same extent in the pancreatic islets as seen after SG alone. The increased GLP-1-producing cells in the pancreatic islets after SG or II-SG were located around the insulin-producing ß cells. CONCLUSIONS: The present study provides evidence supporting the hindgut hypothesis. II-SG increased GLP-1 production both in the interposed ileum and in the pancreatic islets, leading to metabolic beneficial effects and altered eating behavior.


Asunto(s)
Conducta Alimentaria , Gastroplastia/métodos , Péptido 1 Similar al Glucagón/metabolismo , Íleon/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Metabolismo Energético , Íleon/cirugía , Masculino , Ratas , Ratas Sprague-Dawley
20.
PLoS One ; 8(2): e55493, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23405159

RESUMEN

Alterations in checkpoint and DNA repair pathways may provide adaptive mechanisms contributing to acquired drug resistance. Here, we investigated the levels of proteins mediating DNA damage signaling and -repair in RPMI8226 multiple myeloma cells and its Melphalan-resistant derivative 8226-LR5. We observed markedly reduced steady-state levels of DNA glycosylases UNG2, NEIL1 and MPG in the resistant cells and cross-resistance to agents inducing their respective DNA base lesions. Conversely, repair of alkali-labile sites was apparently enhanced in the resistant cells, as substantiated by alkaline comet assay, autoribosylation of PARP-1, and increased sensitivity to PARP-1 inhibition by 4-AN or KU58684. Reduced base-excision and enhanced single-strand break repair would both contribute to the observed reduction in genomic alkali-labile sites, which could jeopardize productive processing of the more cytotoxic Melphalan-induced interstrand DNA crosslinks (ICLs). Furthermore, we found a marked upregulation of proteins in the non-homologous end-joining (NHEJ) pathway of double-strand break (DSB) repair, likely contributing to the observed increase in DSB repair kinetics in the resistant cells. Finally, we observed apparent upregulation of ATR-signaling and downregulation of ATM-signaling in the resistant cells. This was accompanied by markedly increased sensitivity towards Melphalan in the presence of ATR-, DNA-PK, or CHK1/2 inhibitors whereas no sensitizing effect was observed subsequent to ATM inhibition, suggesting that replication blocking lesions are primary triggers of the DNA damage response in the Melphalan resistant cells. In conclusion, Melphalan resistance is apparently contributed by modulation of the DNA damage response at multiple levels, including downregulation of specific repair pathways to avoid repair intermediates that could impair efficient processing of cytotoxic ICLs and ICL-induced DSBs. This study has revealed several novel candidate biomarkers for Melphalan sensitivity that will be included in targeted quantitation studies in larger patient cohorts to validate their value in prognosis as well as targets for replacement- or adjuvant therapies.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Biomarcadores de Tumor/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN/genética , Resistencia a Antineoplásicos/genética , Melfalán/farmacología , Mieloma Múltiple/genética , 8-Hidroxi-2'-Desoxicoguanosina , Apoptosis , Western Blotting , Ciclo Celular/genética , Proliferación Celular , Ensayo Cometa , Reparación del ADN/efectos de los fármacos , Replicación del ADN/genética , Proteína Quinasa Activada por ADN , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/patología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA