Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 136(17): 1956-1967, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-32693407

RESUMEN

Gray platelet syndrome (GPS) is a rare recessive disorder caused by biallelic variants in NBEAL2 and characterized by bleeding symptoms, the absence of platelet α-granules, splenomegaly, and bone marrow (BM) fibrosis. Due to the rarity of GPS, it has been difficult to fully understand the pathogenic processes that lead to these clinical sequelae. To discern the spectrum of pathologic features, we performed a detailed clinical genotypic and phenotypic study of 47 patients with GPS and identified 32 new etiologic variants in NBEAL2. The GPS patient cohort exhibited known phenotypes, including macrothrombocytopenia, BM fibrosis, megakaryocyte emperipolesis of neutrophils, splenomegaly, and elevated serum vitamin B12 levels. Novel clinical phenotypes were also observed, including reduced leukocyte counts and increased presence of autoimmune disease and positive autoantibodies. There were widespread differences in the transcriptome and proteome of GPS platelets, neutrophils, monocytes, and CD4 lymphocytes. Proteins less abundant in these cells were enriched for constituents of granules, supporting a role for Nbeal2 in the function of these organelles across a wide range of blood cells. Proteomic analysis of GPS plasma showed increased levels of proteins associated with inflammation and immune response. One-quarter of plasma proteins increased in GPS are known to be synthesized outside of hematopoietic cells, predominantly in the liver. In summary, our data show that, in addition to the well-described platelet defects in GPS, there are immune defects. The abnormal immune cells may be the drivers of systemic abnormalities such as autoimmune disease.


Asunto(s)
Gránulos Citoplasmáticos/patología , Heterogeneidad Genética , Síndrome de Plaquetas Grises , Sistema Inmunológico/patología , Fenotipo , Biopsia , Proteínas Sanguíneas/genética , Estudios de Casos y Controles , Estudios de Cohortes , Gránulos Citoplasmáticos/metabolismo , Diagnóstico Diferencial , Frecuencia de los Genes , Estudios de Asociación Genética , Síndrome de Plaquetas Grises/clasificación , Síndrome de Plaquetas Grises/genética , Síndrome de Plaquetas Grises/inmunología , Síndrome de Plaquetas Grises/patología , Humanos , Sistema Inmunológico/fisiología , Enfermedades del Sistema Inmune/sangre , Enfermedades del Sistema Inmune/diagnóstico , Enfermedades del Sistema Inmune/genética , Enfermedades del Sistema Inmune/patología , Mutación
2.
Brain ; 144(8): 2427-2442, 2021 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-33792664

RESUMEN

Marinesco-Sjögren syndrome is a rare human disorder caused by biallelic mutations in SIL1 characterized by cataracts in infancy, myopathy and ataxia, symptoms which are also associated with a novel disorder caused by mutations in INPP5K. While these phenotypic similarities may suggest commonalties at a molecular level, an overlapping pathomechanism has not been established yet. In this study, we present six new INPP5K patients and expand the current mutational and phenotypical spectrum of the disease showing the clinical overlap between Marinesco-Sjögren syndrome and the INPP5K phenotype. We applied unbiased proteomic profiling on cells derived from Marinesco-Sjögren syndrome and INPP5K patients and identified alterations in d-3-PHGDH as a common molecular feature. d-3-PHGDH modulates the production of l-serine and mutations in this enzyme were previously associated with a neurological phenotype, which clinically overlaps with Marinesco-Sjögren syndrome and INPP5K disease. As l-serine administration represents a promising therapeutic strategy for d-3-PHGDH patients, we tested the effect of l-serine in generated sil1, phgdh and inpp5k a+b zebrafish models, which showed an improvement in their neuronal phenotype. Thus, our study defines a core phenotypical feature underpinning a key common molecular mechanism in three rare diseases and reveals a common and novel therapeutic target for these patients.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Inositol Polifosfato 5-Fosfatasas/genética , Mutación , Fenotipo , Fosfoglicerato-Deshidrogenasa/genética , Degeneraciones Espinocerebelosas/genética , Adolescente , Adulto , Animales , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Proteómica , Degeneraciones Espinocerebelosas/patología , Pez Cebra
3.
Arch Toxicol ; 96(8): 2341-2360, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35579693

RESUMEN

Remdesivir is a prodrug of a nucleoside analog and the first antiviral therapeutic approved for coronavirus disease. Recent cardiac safety concerns and reports on remdesivir-related acute kidney injury call for a better characterization of remdesivir toxicity and understanding of the underlying mechanisms. Here, we performed an in vitro toxicity assessment of remdesivir around clinically relevant concentrations (Cmax 9 µM) using H9c2 rat cardiomyoblasts, neonatal mouse cardiomyocytes (NMCM), rat NRK-52E and human RPTEC/TERT1 cells as cell models for the assessment of cardiotoxicity or nephrotoxicity, respectively. Due to the known potential of nucleoside analogs for the induction of mitochondrial toxicity, we assessed mitochondrial function in response to remdesivir treatment, early proteomic changes in NMCM and RPTEC/TERT1 cells and the contractile function of NMCM. Short-term treatments (24 h) of H9c2 and NRK-52E cells with remdesivir adversely affected cell viability by inhibition of proliferation as determined by significantly decreased 3H-thymidine uptake. Mitochondrial toxicity of remdesivir (1.6-3.1 µM) in cardiac cells was evident by a significant decrease in oxygen consumption, a collapse of mitochondrial membrane potential and an increase in lactate secretion after a 24-48-h treatment. This was supported by early proteomic changes of respiratory chain proteins and intermediate filaments that are typically involved in mitochondrial reorganization. Functionally, an impedance-based analysis showed that remdesivir (6.25 µM) affected the beat rate and contractility of NMCM. In conclusion, we identified adverse effects of remdesivir in cardiac and kidney cells at clinically relevant concentrations, suggesting a careful evaluation of therapeutic use in patients at risk for cardiovascular or kidney disease.


Asunto(s)
Antivirales , Proteómica , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/análogos & derivados , Animales , Antivirales/toxicidad , Proliferación Celular , Humanos , Riñón , Ratones , Ratas
4.
Plant Physiol ; 184(1): 148-164, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32571844

RESUMEN

Protein homeostasis in eukaryotic organelles and their progenitor prokaryotes is regulated by a series of proteases including the caseinolytic protease (CLPP). CLPP has essential roles in chloroplast biogenesis and maintenance, but the significance of the plant mitochondrial CLPP remains unknown and factors that aid coordination of nuclear- and mitochondrial-encoded subunits for complex assembly in mitochondria await discovery. We generated knockout lines of the single gene for the mitochondrial CLP protease subunit, CLPP2, in Arabidopsis (Arabidopsis thaliana). Mutants showed a higher abundance of transcripts from mitochondrial genes encoding oxidative phosphorylation protein complexes, whereas nuclear genes encoding other subunits of the same complexes showed no change in transcript abundance. By contrast, the protein abundance of specific nuclear-encoded subunits in oxidative phosphorylation complexes I and V increased in CLPP2 knockouts, without accumulation of mitochondrial-encoded counterparts in the same complex. Complexes with subunits mainly or entirely encoded in the nucleus were unaffected. Analysis of protein import and function of complex I revealed that while function was retained, protein homeostasis was disrupted, leading to accumulation of soluble subcomplexes of nuclear-encoded subunits. Therefore, CLPP2 contributes to the mitochondrial protein degradation network through supporting coordination and homeostasis of protein complexes encoded across mitochondrial and nuclear genomes.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Núcleo Celular/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Endopeptidasa Clp/metabolismo , Regulación de la Expresión Génica de las Plantas , Fosforilación Oxidativa
5.
Proteomics ; 20(11): e1900104, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32104967

RESUMEN

Growth differentiation factor 11 (GDF11) is a TGF-ß superfamily circulating factor that regulates cardiomyocyte size in rodents, sharing 90% amino acid sequence identity in the active domains with myostatin (GDF8)-the major determinant of skeletal muscle mass. Conflicting data on age-related changes in circulating levels have been reported mainly due to the lack of specific detection methods. More recently, liquid chromatography tandem mass spectrometry (LC-MS/MS) based assay showed that the circulating levels of GDF11 do not change significantly throughout human lifespan, but GDF8 levels decrease with aging in men. Here a novel detection method is demonstrated based on parallel reaction monitoring LC-MS/MS assay combined with immunoprecipitation to reliably distinguish GDF11 and GDF8 as well as determine their endogenous levels in mouse serum. The data indicate that both GDF11 and GDF8 circulating levels significantly decline with aging in female mice.


Asunto(s)
Proteínas Morfogenéticas Óseas/sangre , Factores de Diferenciación de Crecimiento/sangre , Miostatina/sangre , Envejecimiento/fisiología , Animales , Cromatografía Liquida , Femenino , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Proteómica , Espectrometría de Masas en Tándem
6.
Hum Mutat ; 41(9): 1600-1614, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32516863

RESUMEN

Filamin C (encoded by the FLNC gene) is a large actin-cross-linking protein involved in shaping the actin cytoskeleton in response to signaling events both at the sarcolemma and at myofibrillar Z-discs of cross-striated muscle cells. Multiple mutations in FLNC are associated with myofibrillar myopathies of autosomal-dominant inheritance. Here, we describe for the first time a boy with congenital onset of generalized muscular hypotonia and muscular weakness, delayed motor development but no cardiac involvement associated with a homozygous FLNC mutation c.1325C>G (p.Pro442Arg). We performed ultramorphological, proteomic, and functional investigations as well as immunological studies of known marker proteins for dominant filaminopathies. We show that the mutant protein is expressed in similar quantities as the wild-type variant in control skeletal muscle fibers. The proteomic signature of quadriceps muscle is altered and ultrastructural perturbations are evident. Moreover, filaminopathy marker proteins are comparable both in our homozygous and a dominant control case (c.5161delG). Biochemical investigations demonstrate that the recombinant mutant protein is less stable and more prone to degradation by proteolytic enzymes than the wild-type variant. The unusual congenital presentation of the disease clearly demonstrates that homozygosity for mutations in FLNC severely aggravates the phenotype.


Asunto(s)
Filaminas/genética , Miopatías Estructurales Congénitas/genética , Adolescente , Niño , Preescolar , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Proteoma
7.
Mol Cell Proteomics ; 17(4): 826-834, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29358340

RESUMEN

Proteases are in the center of many diseases, and consequently, proteases and their substrates are important drug targets as represented by an estimated 5-10% of all drugs under development. Mass spectrometry has been an indispensable tool for the discovery of novel protease substrates, particularly through the proteome-scale enrichment of so-called N-terminal peptides representing endogenous protein N termini. Methods such as combined fractional diagonal chromatography (COFRADIC)1 and, later, terminal amine isotopic labeling of substrates (TAILS) have revealed numerous insights into protease substrates and consensus motifs. We present an alternative and simple protocol for N-terminal peptide enrichment, based on charge-based fractional diagonal chromatography (ChaFRADIC) and requiring only well-established protein chemistry and a pipette tip. Using iTRAQ-8-plex, we quantified on average 2,073 ± 52 unique N-terminal peptides from only 4.3 µg per sample/channel, allowing the identification of proteolytic targets and consensus motifs. This high sensitivity may even allow working with clinical samples such as needle biopsies in the future. We applied our method to study the dynamics of staurosporine-induced apoptosis. Our data demonstrate an orchestrated regulation of specific pathways after 1.5 h, 3 h, and 6 h of treatment, with many important players of homeostasis targeted already after 1.5 h. We additionally observed an early multilevel modulation of the splicing machinery both by proteolysis and phosphorylation. This may reflect the known role of alternative splicing variants for a variety of apoptotic genes, which seems to be a driving force of staurosporine-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Péptido Hidrolasas/metabolismo , Proteómica/métodos , Estaurosporina/farmacología , Línea Celular Tumoral , Cromatografía/métodos , Humanos , Espectrometría de Masas/métodos
8.
Proc Natl Acad Sci U S A ; 114(23): E4621-E4630, 2017 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-28536193

RESUMEN

Podocytes form the outer part of the glomerular filter, where they have to withstand enormous transcapillary filtration forces driving glomerular filtration. Detachment of podocytes from the glomerular basement membrane precedes most glomerular diseases. However, little is known about the regulation of podocyte adhesion in vivo. Thus, we systematically screened for podocyte-specific focal adhesome (FA) components, using genetic reporter models in combination with iTRAQ-based mass spectrometry. This approach led to the identification of FERM domain protein EPB41L5 as a highly enriched podocyte-specific FA component in vivo. Genetic deletion of Epb41l5 resulted in severe proteinuria, detachment of podocytes, and development of focal segmental glomerulosclerosis. Remarkably, by binding and recruiting the RhoGEF ARGHEF18 to the leading edge, EPB41L5 directly controls actomyosin contractility and subsequent maturation of focal adhesions, cell spreading, and migration. Furthermore, EPB41L5 controls matrix-dependent outside-in signaling by regulating the focal adhesome composition. Thus, by linking extracellular matrix sensing and signaling, focal adhesion maturation, and actomyosin activation EPB41L5 ensures the mechanical stability required for podocytes at the kidney filtration barrier. Finally, a diminution of EPB41L5-dependent signaling programs appears to be a common theme of podocyte disease, and therefore offers unexpected interventional therapeutic strategies to prevent podocyte loss and kidney disease progression.


Asunto(s)
Actomiosina/metabolismo , Proteínas del Citoesqueleto/metabolismo , Adhesiones Focales/metabolismo , Proteínas de la Membrana/metabolismo , Podocitos/metabolismo , Animales , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Femenino , Adhesiones Focales/patología , Técnicas de Inactivación de Genes , Glomeruloesclerosis Focal y Segmentaria/etiología , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Síndrome Nefrótico/etiología , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/patología , Podocitos/patología , Embarazo , Proteómica , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Transducción de Señal
9.
Am J Physiol Endocrinol Metab ; 317(2): E374-E387, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31211616

RESUMEN

Mitochondria are dynamic organelles with diverse functions in tissues such as liver and skeletal muscle. To unravel the mitochondrial contribution to tissue-specific physiology, we performed a systematic comparison of the mitochondrial proteome and lipidome of mice and assessed the consequences hereof for respiration. Liver and skeletal muscle mitochondrial protein composition was studied by data-independent ultra-high-performance (UHP)LC-MS/MS-proteomics, and lipid profiles were compared by UHPLC-MS/MS lipidomics. Mitochondrial function was investigated by high-resolution respirometry in samples from mice and humans. Enzymes of pyruvate oxidation as well as several subunits of complex I, III, and ATP synthase were more abundant in muscle mitochondria. Muscle mitochondria were enriched in cardiolipins associated with higher oxidative phosphorylation capacity and flexibility, in particular CL(18:2)4 and 22:6-containing cardiolipins. In contrast, protein equipment of liver mitochondria indicated a shuttling of complex I substrates toward gluconeogenesis and ketogenesis and a higher preference for electron transfer via the flavoprotein quinone oxidoreductase pathway. Concordantly, muscle and liver mitochondria showed distinct respiratory substrate preferences. Muscle respired significantly more on the complex I substrates pyruvate and glutamate, whereas in liver maximal respiration was supported by complex II substrate succinate. This was a consistent finding in mouse liver and skeletal muscle mitochondria and human samples. Muscle mitochondria are tailored to produce ATP with a high capacity for complex I-linked substrates. Liver mitochondria are more connected to biosynthetic pathways, preferring fatty acids and succinate for oxidation. The physiologic diversity of mitochondria may help to understand tissue-specific disease pathologies and to develop therapies targeting mitochondrial function.


Asunto(s)
Metabolismo Energético/fisiología , Hígado/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Músculo Esquelético/metabolismo , Proteoma/metabolismo , Animales , Femenino , Humanos , Hígado/química , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Hepáticas/metabolismo , Mitocondrias Musculares/metabolismo , Proteínas Mitocondriales/análisis , Músculo Esquelético/química , Especificidad de Órganos , Mapeo Peptídico/métodos , Proteoma/análisis
10.
Adv Exp Med Biol ; 1158: 143-182, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31452140

RESUMEN

Mitochondrial dysfunction is discussed as a key player in the pathogenesis of type 2 diabetes mellitus (T2Dm), a highly prevalent disease rapidly developing as one of the greatest global health challenges of this century. Data however about the involvement of mitochondria, central hubs in bioenergetic processes, in the disease development are still controversial. Lipid and protein homeostasis are under intense discussion to be crucial for proper mitochondrial function. Consequently proteomics and lipidomics analyses might help to understand how molecular changes in mitochondria translate to alterations in energy transduction as observed in the healthy and metabolic diseases such as T2Dm and other related disorders. Mitochondrial lipids integrated in a tool covering proteomic and functional analyses were up to now rarely investigated, although mitochondrial lipids might provide a possible lynchpin in the understanding of type 2 diabetes development and thereby prevention. In this chapter state-of-the-art analytical strategies, pre-analytical aspects, potential pitfalls as well as current proteomics and lipidomics-based knowledge about the pathophysiological role of mitochondria in the pathogenesis of type 2 diabetes will be discussed.


Asunto(s)
Biología Computacional , Diabetes Mellitus Tipo 2 , Hígado , Mitocondrias , Músculo Esquelético , Proteómica , Diabetes Mellitus Tipo 2/fisiopatología , Humanos , Metabolismo de los Lípidos , Hígado/fisiopatología , Mitocondrias/metabolismo , Músculo Esquelético/fisiopatología
11.
Arch Toxicol ; 92(6): 1939-1952, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29761207

RESUMEN

Bosentan is well known to induce cholestatic liver toxicity in humans. The present study was set up to characterize the hepatotoxic effects of this drug at the transcriptomic, proteomic, and metabolomic levels. For this purpose, human hepatoma-derived HepaRG cells were exposed to a number of concentrations of bosentan during different periods of time. Bosentan was found to functionally and transcriptionally suppress the bile salt export pump as well as to alter bile acid levels. Pathway analysis of both transcriptomics and proteomics data identified cholestasis as a major toxicological event. Transcriptomics results further showed several gene changes related to the activation of the nuclear farnesoid X receptor. Induction of oxidative stress and inflammation were also observed. Metabolomics analysis indicated changes in the abundance of specific endogenous metabolites related to mitochondrial impairment. The outcome of this study may assist in the further optimization of adverse outcome pathway constructs that mechanistically describe the processes involved in cholestatic liver injury.


Asunto(s)
Miembro 11 de la Subfamilia B de Transportador de Casetes de Unión al ATP/metabolismo , Bosentán/toxicidad , Hígado/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Ácidos y Sales Biliares/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Humanos , Hígado/metabolismo , Metabolómica , Estrés Oxidativo/genética , Proteómica , Receptores Citoplasmáticos y Nucleares/genética
12.
BMC Bioinformatics ; 18(1): 148, 2017 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-28253837

RESUMEN

BACKGROUND: The classification of samples on a molecular level has manifold applications, from patient classification regarding cancer treatment to phylogenetics for identifying evolutionary relationships between species. Modern methods employ the alignment of DNA or amino acid sequences, mostly not genome-wide but only on selected parts of the genome. Recently proteomics-based approaches have become popular. An established method for the identification of peptides and proteins is liquid chromatography-tandem mass spectrometry (LC-MS/MS). First, protein sequences from MS/MS spectra are identified by means of database searches, given samples with known genome-wide sequence information, then sequence based methods are applied. Alternatively, de novo peptide sequencing algorithms annotate MS/MS spectra and deduce peptide/protein information without a database. A newer approach independent of additional information is to directly compare unidentified tandem mass spectra. The challenge then is to compute the distance between pairwise MS/MS runs consisting of thousands of spectra. METHODS: We present DISMS2, a new algorithm to calculate proteome-wide distances directly from MS/MS data, extending the algorithm compareMS2, an approach that also uses a spectral comparison pipeline. RESULTS: Our new more flexible algorithm, DISMS2, allows for the choice of the spectrum distance measure and includes different spectra preprocessing and filtering steps that can be tailored to specific situations by parameter optimization. CONCLUSIONS: DISMS2 performs well for samples from species with and without database annotation and thus has clear advantages over methods that are purely based on database search.


Asunto(s)
Algoritmos , Cromatografía Liquida/métodos , Péptidos/análisis , Proteoma/química , Proteómica/métodos , Espectrometría de Masas en Tándem/métodos , Secuencia de Aminoácidos , Bases de Datos de Proteínas , Humanos
13.
J Proteome Res ; 16(9): 3209-3218, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28741358

RESUMEN

Complex mass spectrometry based proteomics data sets are mostly analyzed by protein database searches. While this approach performs considerably well for sequenced organisms, direct inference of peptide sequences from tandem mass spectra, i.e., de novo peptide sequencing, oftentimes is the only way to obtain information when protein databases are absent. However, available algorithms suffer from drawbacks such as lack of validation and often high rates of false positive hits (FP). Here we present a simple method of combining results from commonly available de novo peptide sequencing algorithms, which in conjunction with minor tweaks in data acquisition ensues lower empirical FDR compared to the analysis using single algorithms. Results were validated using state-of-the art database search algorithms as well specifically synthesized reference peptides. Thus, we could increase the number of PSMs meeting a stringent FDR of 5% more than 3-fold compared to the single best de novo sequencing algorithm alone, accounting for an average of 11 120 PSMs (combined) instead of 3476 PSMs (alone) in triplicate 2 h LC-MS runs of tryptic HeLa digestion.


Asunto(s)
Algoritmos , Péptidos/análisis , Proteómica/métodos , Análisis de Secuencia de Proteína/métodos , Secuencia de Aminoácidos , Animales , Línea Celular , Cromatografía Liquida , Bases de Datos de Proteínas , Células HeLa , Humanos , Ratones , Mioblastos/química , Mioblastos/metabolismo , Proteolisis , Proteómica/instrumentación , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Caracoles/química , Caracoles/metabolismo , Espectrometría de Masas en Tándem , Tripsina/química
14.
J Biol Chem ; 291(44): 23330-23342, 2016 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-27645995

RESUMEN

In the chloroplast of the green alga Chlamydomonas reinhardtii, two discontinuous group II introns, psaA-i1 and psaA-i2, splice in trans, and thus their excision process resembles the nuclear spliceosomal splicing pathway. Here, we address the question whether fragmentation of trans-acting RNAs is accompanied by the formation of a chloroplast spliceosome-like machinery. Using a combination of liquid chromatography-mass spectrometry (LC-MS), size exclusion chromatography, and quantitative RT-PCR, we provide the first characterization of a high molecular weight ribonucleoprotein apparatus participating in psaA mRNA splicing. This supercomplex contains two subcomplexes (I and II) that are responsible for trans-splicing of either psaA-i1 or psaA-i2. We further demonstrate that both subcomplexes are associated with intron RNA, which is a prerequisite for the correct assembly of subcomplex I. This study contributes further to our view of how the eukaryotic nuclear spliceosome evolved after bacterial endosymbiosis through fragmentation of self-splicing group II introns into a dynamic, protein-rich RNP machinery.


Asunto(s)
Chlamydomonas reinhardtii/genética , Cloroplastos/metabolismo , Intrones , Ribonucleoproteínas/metabolismo , Trans-Empalme , Chlamydomonas reinhardtii/metabolismo , Cloroplastos/genética , Ribonucleoproteínas/genética , Empalmosomas/genética , Empalmosomas/metabolismo
15.
Plant J ; 85(1): 57-69, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26611495

RESUMEN

Splicing of organellar introns requires the activity of numerous nucleus-encoded factors. In the chloroplast of Chlamydomonas reinhardtii, maturation of psaA mRNA encoding photosystem I subunit A involves two steps of trans-splicing. The exons, located on three separate transcripts, are flanked by sequences that fold to form the conserved structures of two group II introns. A fourth transcript contributes to assembly of the first intron, which is thus tripartite. The raa7 mutant (RNA maturation of psaA 7) is deficient in trans-splicing of the second intron of psaA, and may be rescued by transforming the chloroplast genome with an intron-less version of psaA. Using mapped-based cloning, we identify the RAA7 locus, which encodes a pioneer protein with no previously known protein domain or motif. The Raa7 protein, which is not associated with membranes, localizes to the chloroplast. Raa7 is a component of a large complex and co-sediments in sucrose gradients with the previously described splicing factors Raa1 and Raa2. Based on tandem affinity purification of Raa7 and mass spectrometry, Raa1 and Raa2 were identified as interacting partners of Raa7. Yeast two-hybrid experiments indicate that the interaction of Raa7 with Raa1 and Raa2 may be direct. We conclude that Raa7 is a component of a multimeric complex that is required for trans-splicing of the second intron of psaA. The characterization of this psaA trans-splicing complex is also of interest from an evolutionary perspective because the nuclear spliceosomal introns are thought to derive from group II introns, with which they show mechanistic and structural similarity.


Asunto(s)
Chlamydomonas reinhardtii/genética , Trans-Empalme/genética , Núcleo Celular/metabolismo , Cloroplastos/metabolismo , Exones/genética , Intrones , Complejo de Proteína del Fotosistema I/metabolismo , Dominios Proteicos , Empalme del ARN , ARN Mensajero/genética , Empalmosomas/metabolismo
16.
J Proteome Res ; 15(3): 945-55, 2016 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-26781476

RESUMEN

Studying (neuro)muscular disorders is a major topic in biomedicine with a demand for suitable model systems. Continuous cell culture (in vitro) systems have several technical advantages over in vivo systems and became widely used tools for discovering physiological/pathophysiological mechanisms in muscle. In particular, myoblast cell lines are suitable model systems to study complex biochemical adaptations occurring in skeletal muscle and cellular responses to altered genetic/environmental conditions. Whereas most in vitro studies use extensively characterized murine C2C12 cells, a comprehensive description of an equivalent human cell line, not genetically manipulated for immortalization, is lacking. Therefore, we characterized human immortal myoblastic RCMH cells using scanning (SEM) and transmission electron microscopy (TEM) and proteomics. Among more than 6200 identified proteins we confirm the known expression of proteins important for muscle function. Comparing the RCMH proteome with two well-defined nonskeletal muscle cells lines (HeLa, U2OS) revealed a considerable enrichment of proteins important for muscle function. SEM/TEM confirmed the presence of agglomerates of cytoskeletal components/intermediate filaments and a prominent rough ER. In conclusion, our results indicate RMCH as a suitable in vitro model for investigating muscle function-related processes such as mechanical stress burden and mechanotransduction, EC coupling, cytoskeleton, muscle cell metabolism and development, and (ER-associated) myopathic disorders.


Asunto(s)
Mioblastos/metabolismo , Proteoma/metabolismo , Línea Celular , Retículo Endoplásmico/patología , Humanos , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Mioblastos/patología
17.
Arch Toxicol ; 90(6): 1529-1539, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27129694

RESUMEN

SEURAT-1 is a joint research initiative between the European Commission and Cosmetics Europe aiming to develop in vitro- and in silico-based methods to replace the in vivo repeated dose systemic toxicity test used for the assessment of human safety. As one of the building blocks of SEURAT-1, the DETECTIVE project focused on a key element on which in vitro toxicity testing relies: the development of robust and reliable, sensitive and specific in vitro biomarkers and surrogate endpoints that can be used for safety assessments of chronically acting toxicants, relevant for humans. The work conducted by the DETECTIVE consortium partners has established a screening pipeline of functional and "-omics" technologies, including high-content and high-throughput screening platforms, to develop and investigate human biomarkers for repeated dose toxicity in cellular in vitro models. Identification and statistical selection of highly predictive biomarkers in a pathway- and evidence-based approach constitute a major step in an integrated approach towards the replacement of animal testing in human safety assessment. To discuss the final outcomes and achievements of the consortium, a meeting was organized in Brussels. This meeting brought together data-producing and supporting consortium partners. The presentations focused on the current state of ongoing and concluding projects and the strategies employed to identify new relevant biomarkers of toxicity. The outcomes and deliverables, including the dissemination of results in data-rich "-omics" databases, were discussed as were the future perspectives of the work completed under the DETECTIVE project. Although some projects were still in progress and required continued data analysis, this report summarizes the presentations, discussions and the outcomes of the project.


Asunto(s)
Alternativas a las Pruebas en Animales/métodos , Pruebas de Toxicidad/métodos , Alternativas a las Pruebas en Animales/legislación & jurisprudencia , Alternativas a las Pruebas en Animales/organización & administración , Animales , Biomarcadores/análisis , Células Cultivadas , Seguridad de Productos para el Consumidor , Unión Europea , Regulación Gubernamental , Ensayos Analíticos de Alto Rendimiento , Humanos , Técnicas In Vitro
18.
Angew Chem Int Ed Engl ; 55(36): 10909-12, 2016 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-27496389

RESUMEN

Targeting acquired drug resistance represents the major challenge in the treatment of EGFR-driven non-small-cell lung cancer (NSCLC). Herein, we describe the structure-based design, synthesis, and biological evaluation of a novel class of covalent EGFR inhibitors that exhibit excellent inhibition of EGFR-mutant drug-resistant cells. Protein X-ray crystallography combined with detailed kinetic studies led to a deeper understanding of the mode of inhibition of EGFR-T790M and provided insight into the key principles for effective inhibition of the recently discovered tertiary mutation at EGFR-C797S.


Asunto(s)
Receptores ErbB/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Sitios de Unión , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Humanos , Cinética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Simulación del Acoplamiento Molecular , Fosforilación , Mutación Puntual , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/química , Pirazoles/metabolismo , Pirazoles/farmacología , Piridinas/química , Piridinas/metabolismo , Piridinas/farmacología
19.
Proteomics ; 14(4-5): 513-24, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24339426

RESUMEN

Beside gene expression and translational control, which are relatively slow, PTM of proteins represents the major level of regulation, from very fast and reversible to slow or irreversible processes. PTMs affect protein structure and act as molecular switches, which regulate the interaction of proteins with DNA, cofactors, lipids, and other proteins. In the past few years, evidence for extensive crosstalk between PTMs has accumulated. The combination of different PTMs on protein surfaces can create a "PTM code," which can be recognized by specific effectors to initiate/inhibit downstream events, only inducing/retaining a signal once the complementary incoming signals are present at the same time and place. Although MS-based proteomics has substantially improved our knowledge about PTMs, currently sensitive and dedicated analytical strategies are available only for few different types of PTM. Several recent studies focused on the combinatorial analysis of PTMs, but preferentially utilized peptide-centric bottom-up strategies might be too restricted to decipher complex PTM codes. Here, we discuss the current state of PTM crosstalk research and how proteomics may contribute to understanding PTM codes, representing the next level of complexity and one of the biggest challenges for future proteomics research.


Asunto(s)
Espectrometría de Masas/métodos , Procesamiento Proteico-Postraduccional/genética , Proteínas/metabolismo , Proteómica/métodos , Secuencia de Aminoácidos , Humanos , Fosforilación/genética , Proteínas/genética
20.
J Biol Chem ; 288(49): 35237-52, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24158441

RESUMEN

In metazoans, the highly conserved MAPK signaling pathway regulates cell fate decision. Aberrant activation of this pathway has been implicated in multiple human cancers and some developmental disorders. KSR1 functions as an essential scaffold that binds the individual components of the cascade and coordinates their assembly into multiprotein signaling platforms. The mechanism of KSR1 regulation is highly complex and not completely understood. In this study, we identified Tyr(728) as a novel regulatory phosphorylation site in KSR1. We show that Tyr(728) is phosphorylated by LCK, uncovering an additional and unexpected link between Src kinases and MAPK signaling. To understand how phosphorylation of Tyr(728) may regulate the role of KSR1 in signal transduction, we integrated structural modeling and biochemical studies. We demonstrate that Tyr(728) is involved in maintaining the conformation of the KSR1 kinase domain required for binding to MEK. It also affects phosphorylation and activation of MEK by RAF kinases and consequently influences cell proliferation. Moreover, our studies suggest that phosphorylation of Tyr(728) may affect the intrinsic kinase activity of KSR1. Together, we propose that phosphorylation of Tyr(728) may regulate the transition between the scaffolding and the catalytic function of KSR1 serving as a control point used to fine-tune cellular responses.


Asunto(s)
Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Proliferación Celular , Activación Enzimática , Humanos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Fosforilación , Unión Proteica , Conformación Proteica , Proteínas Quinasas/genética , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Tirosina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA