Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Biol ; 21(1): 164, 2023 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-37525144

RESUMEN

BACKGROUND: Individual functional modifications shape the ability of wildlife populations to cope with anthropogenic environmental changes. But instead of adaptive response, human-altered environments can generate a succession of deleterious functional changes leading to the extinction of the population. To study how persistent anthropogenic changes impacted local species' population status, we characterised population structure, genetic diversity and individual response of gene expression in the tree frog Hyla orientalis along a gradient of radioactive contamination around the Chernobyl nuclear power plant. RESULTS: We detected lower effective population size in populations most exposed to ionizing radiation in the Chernobyl Exclusion Zone that is not compensated by migrations from surrounding areas. We also highlighted a decreased body condition of frogs living in the most contaminated area, a distinctive transcriptomics signature and stop-gained mutations in genes involved in energy metabolism. While the association with dose will remain correlational until further experiments, a body of evidence suggests the direct or indirect involvement of radiation exposure in these changes. CONCLUSIONS: Despite ongoing migration and lower total dose rates absorbed than at the time of the accident, our results demonstrate that Hyla orientalis specimens living in the Chernobyl Exclusion Zone are still undergoing deleterious changes, emphasizing the long-term impacts of the nuclear disaster.


Asunto(s)
Accidente Nuclear de Chernóbil , Animales , Humanos , Densidad de Población , Animales Salvajes , Radiación Ionizante , Anuros/genética
2.
Mol Ther ; 31(2): 471-486, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35965411

RESUMEN

The heat shock protein 27 (Hsp27) has emerged as a principal factor of the castration-resistant prostate cancer (CRPC) progression. Also, an antisense oligonucleotide (ASO) against Hsp27 (OGX-427 or apatorsen) has been assessed in different clinical trials. Here, we illustrate that Hsp27 highly regulates the expression of the human DEAD-box protein 5 (DDX5), and we define DDX5 as a novel therapeutic target for CRPC treatment. DDX5 overexpression is strongly correlated with aggressive tumor features, notably with CRPC. DDX5 downregulation using a specific ASO-based inhibitor that acts on DDX5 mRNAs inhibits cell proliferation in preclinical models, and it particularly restores the treatment sensitivity of CRPC. Interestingly, through the identification and analysis of DDX5 protein interaction networks, we have identified some specific functions of DDX5 in CRPC that could contribute actively to tumor progression and therapeutic resistance. We first present the interactions of DDX5 and the Ku70/80 heterodimer and the transcription factor IIH, thereby uncovering DDX5 roles in different DNA repair pathways. Collectively, our study highlights critical functions of DDX5 contributing to CRPC progression and provides preclinical proof of concept that a combination of ASO-directed DDX5 inhibition with a DNA damage-inducing therapy can serve as a highly potential novel strategy to treat CRPC.


Asunto(s)
Oligonucleótidos Antisentido , Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/uso terapéutico , Oligonucleótidos Antisentido/farmacología , Neoplasias de la Próstata Resistentes a la Castración/terapia , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , ARN Mensajero/uso terapéutico , Proteínas de Choque Térmico HSP27/genética , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas de Choque Térmico HSP27/uso terapéutico , Línea Celular Tumoral , ARN Helicasas DEAD-box/genética
3.
Nucleic Acids Res ; 50(5): 2667-2680, 2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35166826

RESUMEN

The tumour suppressor SLX4 plays multiple roles in the maintenance of genome stability, acting as a scaffold for structure-specific endonucleases and other DNA repair proteins. It directly interacts with the mismatch repair (MMR) protein MSH2 but the significance of this interaction remained unknown until recent findings showing that MutSß (MSH2-MSH3) stimulates in vitro the SLX4-dependent Holliday junction resolvase activity. Here, we characterize the mode of interaction between SLX4 and MSH2, which relies on an MSH2-interacting peptide (SHIP box) that drives interaction of SLX4 with both MutSß and MutSα (MSH2-MSH6). While we show that this MSH2 binding domain is dispensable for the well-established role of SLX4 in interstrand crosslink repair, we find that it mediates inhibition of MutSα-dependent MMR by SLX4, unravelling an unanticipated function of SLX4.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Proteínas de Unión al ADN , Endonucleasas , Proteína 2 Homóloga a MutS , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Proteína 2 Homóloga a MutS/metabolismo
4.
Sci Rep ; 10(1): 6787, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32321993

RESUMEN

Metastatic progression is the leading cause of mortality in breast cancer. Invasive tumor cells develop invadopodia to travel through basement membranes and the interstitial matrix. Substantial efforts have been made to characterize invadopodia molecular composition. However, their full molecular identity is still missing due to the difficulty in isolating them. To fill this gap, we developed a non-hypothesis driven proteomic approach based on the BioID proximity biotinylation technology, using the invadopodia-specific protein Tks5α fused to the promiscuous biotin ligase BirA* as bait. In invasive breast cancer cells, Tks5α fusion concentrated to invadopodia and selectively biotinylated invadopodia components, in contrast to a fusion which lacked the membrane-targeting PX domain (Tks5ß). Biotinylated proteins were isolated by affinity capture and identified by mass spectrometry. We identified known invadopodia components, revealing the pertinence of our strategy. Furthermore, we observed that Tks5 newly identified close neighbors belonged to a biologically relevant network centered on actin cytoskeleton organization. Analysis of Tks5ß interactome demonstrated that some partners bound Tks5 before its recruitment to invadopodia. Thus, the present strategy allowed us to identify novel Tks5 partners that were not identified by traditional approaches and could help get a more comprehensive picture of invadopodia molecular landscape.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Biotinilación/métodos , Podosomas/metabolismo , Proteómica/métodos , Proteínas Adaptadoras del Transporte Vesicular/genética , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/metabolismo , Línea Celular Tumoral , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Espectrometría de Masas/métodos , Unión Proteica , Mapas de Interacción de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
5.
Sci Rep ; 9(1): 20241, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31882844

RESUMEN

Contamination of the environment after the Chernobyl and Fukushima Daiichi nuclear power plant (NPP) disasters led to the exposure of a large number of humans and wild animals to radioactive substances. However, the sub-lethal consequences induced by these absorbed radiological doses remain understudied and the long-term biological impacts largely unknown. We assessed the biological effects of chronic exposure to ionizing radiation (IR) on embryonic development by exposing zebrafish embryo from fertilization and up to 120 hours post-fertilization (hpf) at dose rates of 0.5 mGy/h, 5 mGy/h and 50 mGy/h, thereby encompassing the field of low dose rates defined at 6 mGy/h. Chronic exposure to IR altered larval behaviour in a light-dark locomotor test and affected cardiac activity at a dose rate as low as 0.5 mGy/h. The multi-omics analysis of transcriptome, proteome and transcription factor binding sites in the promoters of the deregulated genes, collectively points towards perturbations of neurogenesis, muscle development, and retinoic acid (RA) signaling after chronic exposure to IR. Whole-mount RNA in situ hybridization confirmed the impaired expression of the transcription factors her4.4 in the central nervous system and myogenin in the developing muscles of exposed embryos. At the organ level, the assessment of muscle histology by transmission electron microscopy (TEM) demonstrated myofibers disruption and altered neuromuscular junctions in exposed larvae at 5 mGy/h and 50 mGy/h. The integration of these multi-level data demonstrates that chronic exposure to low dose rates of IR has an impact on neuronal and muscle progenitor cells, that could lead to motility defects in free swimming larvae at 120 hpf. The mechanistic understanding of these effects allows us to propose a model where deregulation of RA signaling by chronic exposure to IR has pleiotropic effects on neurogenesis and muscle development.


Asunto(s)
Desarrollo Embrionario/efectos de la radiación , Desarrollo de Músculos/efectos de la radiación , Músculos/efectos de la radiación , Sistema Nervioso/efectos de la radiación , Radiación Ionizante , Biología de Sistemas/métodos , Animales , Antineoplásicos/farmacología , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Larva/efectos de los fármacos , Larva/genética , Larva/efectos de la radiación , Desarrollo de Músculos/efectos de los fármacos , Desarrollo de Músculos/genética , Músculos/efectos de los fármacos , Músculos/embriología , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/embriología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/efectos de los fármacos , Transcriptoma/efectos de la radiación , Tretinoina/farmacología , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
6.
mBio ; 10(5)2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31641084

RESUMEN

Respiration is a fundamental process that has to optimally respond to metabolic demand and environmental changes. We previously showed that nitrate respiration, crucial for gut colonization by enterobacteria, is controlled by polar clustering of the nitrate reductase increasing the electron flux through the complex. Here, we show that the formate dehydrogenase electron-donating complex, FdnGHI, also clusters at the cell poles under nitrate-respiring conditions. Its proximity to the nitrate reductase complex was confirmed by its identification in the interactome of the latter, which appears to be specific to the nitrate-respiring condition. Interestingly, we have identified a multiprotein complex dedicated to handle nitric oxide resulting from the enhanced activity of the electron transport chain terminated by nitrate reductase. We demonstrated that the cytoplasmic NADH-dependent nitrite reductase NirBD and the hybrid cluster protein Hcp are key contributors to regulation of the nitric oxide level during nitrate respiration. Thus, gathering of actors involved in respiration and NO homeostasis seems to be critical to balancing maximization of electron flux and the resulting toxicity.IMPORTANCE Most bacteria rely on the redox activity of respiratory complexes embedded in the cytoplasmic membrane to gain energy in the form of ATP and of an electrochemical gradient established across the membrane. Nevertheless, production of harmful and toxic nitric oxide by actively growing bacteria as either an intermediate or side-product of nitrate respiration challenges how homeostasis control is exerted. Here, we show that components of the nitrate electron transport chain are clustered, likely influencing the kinetics of the process. Nitric oxide production from this respiratory chain is controlled and handled through a multiprotein complex, including detoxifying systems. These findings point to an essential role of compartmentalization of respiratory components in bacterial cell growth.


Asunto(s)
Escherichia coli/metabolismo , Nitratos/metabolismo , Membrana Celular/metabolismo , Respiración de la Célula/fisiología , Microscopía Fluorescente , Óxido Nítrico/metabolismo
7.
Proteomics ; 19(21-22): e1800487, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31513346

RESUMEN

SCRIB is a scaffold protein containing leucine-rich repeats (LRR) and PSD-95/Dlg-A/ZO-1 domains (PDZ) that localizes at the basolateral membranes of polarized epithelial cells. Deregulation of its expression or localization leads to epithelial defects and tumorigenesis in part as a consequence of its repressive role on several signaling pathways including AKT, ERK, and HIPPO. In the present work, a proteomic approach is used to characterize the protein complexes associated to SCRIB and its paralogue LANO. Common and specific sets of proteins associated to SCRIB and LANO by MS are identified and an extensive landscape of their associated networks and the first comparative analysis of their respective interactomes are provided. Under proteasome inhibition, it is further found that SCRIB is associated to the ß-catenin destruction complex that is central in Wnt/ß-catenin signaling, a conserved pathway regulating embryonic development and cancer progression. It is shown that the SCRIB/ß-catenin interaction is potentiated upon Wnt3a stimulation and that SCRIB plays a repressing role on Wnt signaling. The data thus provide evidence for the importance of SCRIB in the regulation of the Wnt/ß-catenin pathway.


Asunto(s)
Proteínas Portadoras/genética , Proteínas de la Membrana/genética , Neoplasias/genética , Proteómica , Proteínas Supresoras de Tumor/genética , Carcinogénesis/efectos de los fármacos , Línea Celular Tumoral , Polaridad Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Inhibidores de Proteasoma/farmacología , Transducción de Señal/genética , Vía de Señalización Wnt/genética , Proteína Wnt3A/genética , beta Catenina/genética
8.
Oncotarget ; 9(73): 33762-33777, 2018 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-30333908

RESUMEN

INTRODUCTION: treatments targeting the Human Epidermal Growth Factor Receptor 2 (HER2/ERBB2) have improved the natural history of HER2-positive breast cancer. However, except HER2 protein expression and gene amplification, there is no predictive biomarker to guide the HER2-targeted therapies. We developed Parallel reaction monitoring (PRM) a powerful approach, to quantify and evaluate key proteins involved in the HER2 pathway and/or anti-HER2 treatment sensitivity. RESULTS: in BCLs, PRM measurements correlated with western blot immunocytochemistry and transcriptomic data. At baseline, higher expression of HER2, EGFR, PTEN and HER3 but lower expression of phospho-HER2 correlated with trastuzumab sensitivity. Under trastuzumab, PRM demonstrated a decrease in HER2 and an increase in phospho-HER2, which correlated with drug sensitivity. The opposite was observed under lapatinib. HER2 quantification was also correlated with immunohistochemistry in PDXs and clinical breast cancer samples. DISCUSSION: in conclusion, PRM-based assay, developed to quantify proteins of the HER2 pathway in breast cancer samples revealed a large magnitude of expression, which may have relevance in terms of treatment sensitivity. MATERIALS AND METHODS: we first evaluated PRM in term of sensitivity, linearity and reproducibility. PRM was then applied to breast cancer cell lines (BCLs) including BCLs exposed to anti-HER2 agents, patient-derived xenografts (PDXs) and frozen breast cancer samples.

9.
Proc Natl Acad Sci U S A ; 114(50): E10687-E10696, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29162697

RESUMEN

Control of microtubule dynamics underlies several fundamental processes such as cell polarity, cell division, and cell motility. To gain insights into the mechanisms that control microtubule dynamics during cell motility, we investigated the interactome of the microtubule plus-end-binding protein end-binding 1 (EB1). Via molecular mapping and cross-linking mass spectrometry we identified and characterized a large complex associating a specific isoform of myomegalin termed "SMYLE" (for short myomegalin-like EB1 binding protein), the PKA scaffolding protein AKAP9, and the pericentrosomal protein CDK5RAP2. SMYLE was associated through an evolutionarily conserved N-terminal domain with AKAP9, which in turn was anchored at the centrosome via CDK5RAP2. SMYLE connected the pericentrosomal complex to the microtubule-nucleating complex (γ-TuRC) via Galectin-3-binding protein. SMYLE associated with nascent centrosomal microtubules to promote microtubule assembly and acetylation. Disruption of SMYLE interaction with EB1 or AKAP9 prevented microtubule nucleation and their stabilization at the leading edge of migrating cells. In addition, SMYLE depletion led to defective astral microtubules and abnormal orientation of the mitotic spindle and triggered G1 cell-cycle arrest, which might be due to defective centrosome integrity. As a consequence, SMYLE loss of function had a profound impact on tumor cell motility and proliferation, suggesting that SMYLE might be an important player in tumor progression.


Asunto(s)
Centrosoma/metabolismo , Microtúbulos/metabolismo , Proteínas Musculares/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Sitios de Unión , Proteínas de Ciclo Celular , Proliferación Celular , Proteínas del Citoesqueleto/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Musculares/química , Proteínas Musculares/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Unión Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
10.
Nat Commun ; 8(1): 1420, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-29127277

RESUMEN

Masitinib, a highly selective protein kinase inhibitor, can sensitise gemcitabine-refractory cancer cell lines when used in combination with gemcitabine. Here we report a reverse proteomic approach that identifies the target responsible for this sensitisation: the deoxycytidine kinase (dCK). Masitinib, as well as other protein kinase inhibitors, such as imatinib, interact with dCK and provoke an unforeseen conformational-dependent activation of this nucleoside kinase, modulating phosphorylation of nucleoside analogue drugs. This phenomenon leads to an increase of prodrug phosphorylation of most of the chemotherapeutic drugs activated by this nucleoside kinase. The unforeseen dual activity of protein kinase inhibition/nucleoside kinase activation could be of great therapeutic benefit, through either reducing toxicity of therapeutic agents by maintaining effectiveness at lower doses or by counteracting drug resistance initiated via down modulation of dCK target.


Asunto(s)
Desoxicitidina Quinasa/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Tiazoles/farmacología , Células A549 , Antineoplásicos/química , Antineoplásicos/farmacología , Benzamidas , Línea Celular Tumoral , Cristalografía por Rayos X , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina Quinasa/química , Diseño de Fármacos , Resistencia a Antineoplásicos , Activación Enzimática/efectos de los fármacos , Humanos , Mesilato de Imatinib/química , Mesilato de Imatinib/farmacología , Modelos Biológicos , Modelos Moleculares , Fosforilación , Piperidinas , Polifarmacología , Inhibidores de Proteínas Quinasas/química , Proteómica , Piridinas , Tiazoles/química , Gemcitabina
11.
Nat Commun ; 8: 14105, 2017 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-28102238

RESUMEN

Functional interplay between tumour cells and their neoplastic extracellular matrix plays a decisive role in malignant progression of carcinomas. Here we provide a comprehensive data set of the human HNSCC-associated fibroblast matrisome. Although much attention has been paid to the deposit of collagen, we identify oncofetal fibronectin (FN) as a major and obligate component of the matrix assembled by stromal fibroblasts from head and neck squamous cell carcinomas (HNSCC). FN overexpression in tumours from 435 patients corresponds to an independent unfavourable prognostic indicator. We show that migration of carcinoma collectives on fibrillar FN-rich matrices is achieved through αvß6 and α9ß1 engagement, rather than α5ß1. Moreover, αvß6-driven migration occurs independently of latent TGF-ß activation and Smad-dependent signalling in tumour epithelial cells. These results provide insights into the adhesion-dependent events at the tumour-stroma interface that govern the collective mode of migration adopted by carcinoma cells to invade surrounding stroma in HNSCC.


Asunto(s)
Carcinoma de Células Escamosas , Movimiento Celular/efectos de los fármacos , Fibronectinas/metabolismo , Neoplasias de Cabeza y Cuello , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Movimiento Celular/fisiología , Matriz Extracelular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Integrinas/genética , Integrinas/metabolismo , Masculino , Persona de Mediana Edad , Carcinoma de Células Escamosas de Cabeza y Cuello
12.
PLoS One ; 12(1): e0169988, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28099510

RESUMEN

NUB1 (Nedd8 ultimate buster 1) is an adaptor protein which negatively regulates the ubiquitin-like protein Nedd8 as well as neddylated proteins levels through proteasomal degradation. However, molecular mechanisms underlying this function are not completely understood. Here, we report that the oncogenic E3 ubiquitin ligase Mdm2 is a new NUB1 interacting protein which induces its ubiquitination. Interestingly, we found that Mdm2-mediated ubiquitination of NUB1 is not a proteolytic signal. Instead of promoting the conjugation of polyubiquitin chains and the subsequent proteasomal degradation of NUB1, Mdm2 rather induces its di-ubiquitination on lysine 159. Importantly, mutation of lysine 159 into arginine inhibits NUB1 activity by impairing its negative regulation of Nedd8 and of neddylated proteins. We conclude that Mdm2 acts as a positive regulator of NUB1 function, by modulating NUB1 ubiquitination on lysine 159.


Asunto(s)
Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Células HEK293 , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Lisina/metabolismo , Proteína NEDD8 , Proteínas Proto-Oncogénicas c-mdm2/genética , Factores de Transcripción/genética , Ubiquitinación , Ubiquitinas/genética , Ubiquitinas/metabolismo
13.
Expert Rev Proteomics ; 14(1): 43-54, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27813428

RESUMEN

INTRODUCTION: Breast cancer (BC) is the most common female cancer in the world and was recently deconstructed in different molecular entities. Although most of the recent assays to characterize tumors at the molecular level are genomic-based, proteins are the actual executors of cellular functions and represent the vast majority of targets for anticancer drugs. Accumulated data has demonstrated an important level of quantitative and qualitative discrepancies between genomic/transcriptomic alterations and their protein counterparts, mostly related to the large number of post-translational modifications. Areas covered: This review will present novel proteomics technologies such as Reverse Phase Protein Array (RPPA) or mass-spectrometry (MS) based approaches that have emerged and that could progressively replace old-fashioned methods (e.g. immunohistochemistry, ELISA, etc.) to validate proteins as diagnostic, prognostic or predictive biomarkers, and eventually monitor them in the routine practice. Expert commentary: These different targeted proteomic approaches, able to complement genomic data in BC and characterize tumors more precisely, will permit to go through a more personalized treatment for each patient and tumor.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Proteómica , Transcriptoma/genética , Neoplasias de la Mama/patología , Femenino , Genómica , Humanos , Espectrometría de Masas
14.
J Exp Med ; 213(11): 2437-2457, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27647348

RESUMEN

The RLTPR cytosolic protein, also known as CARMIL2, is essential for CD28 co-stimulation in mice, but its importance in human T cells and mode of action remain elusive. Here, using affinity purification followed by mass spectrometry analysis, we showed that RLTPR acts as a scaffold, bridging CD28 to the CARD11/CARMA1 cytosolic adaptor and to the NF-κB signaling pathway, and identified proteins not found before within the CD28 signaling pathway. We further demonstrated that RLTPR is essential for CD28 co-stimulation in human T cells and that its noncanonical pleckstrin-homology domain, leucine-rich repeat domain, and proline-rich region were mandatory for that task. Although RLTPR is thought to function as an actin-uncapping protein, this property was dispensable for CD28 co-stimulation in both mouse and human. Our findings suggest that the scaffolding role of RLTPR predominates during CD28 co-stimulation and underpins the similar function of RLTPR in human and mouse T cells. Along that line, the lack of functional RLTPR molecules impeded the differentiation toward Th1 and Th17 fates of both human and mouse CD4+ T cells. RLTPR was also expressed in both human and mouse B cells. In the mouse, RLTPR did not play, however, any detectable role in BCR-mediated signaling and T cell-independent B cell responses.


Asunto(s)
Antígenos CD28/metabolismo , Proteínas de Microfilamentos/metabolismo , Linfocitos T/metabolismo , Secuencias de Aminoácidos , Animales , Células Dendríticas/metabolismo , Endocitosis , Marcación de Gen , Células HEK293 , Humanos , Células Jurkat , Células Asesinas Naturales/metabolismo , Linfocitos/metabolismo , Ratones , Proteínas de Microfilamentos/química , Modelos Biológicos , Mutación/genética , Células Mieloides/metabolismo , Dominios Proteicos , Mapeo de Interacción de Proteínas , Multimerización de Proteína , Proteómica , Transducción de Señal , Linfocitos T Reguladores/metabolismo , Timocitos/metabolismo
15.
Hum Mol Genet ; 25(3): 497-513, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26643951

RESUMEN

Oral-facial-digital (OFD) syndromes are rare heterogeneous disorders characterized by the association of abnormalities of the face, the oral cavity and the extremities, some due to mutations in proteins of the transition zone of the primary cilia or the closely associated distal end of centrioles. These two structures are essential for the formation of functional cilia, and for signaling events during development. We report here causal compound heterozygous mutations of KIAA0753/OFIP in a patient with an OFD VI syndrome. We show that the KIAA0753/OFIP protein, whose sequence is conserved in ciliated species, associates with centrosome/centriole and pericentriolar satellites in human cells and forms a complex with FOR20 and OFD1. The decreased expression of any component of this ternary complex in RPE1 cells causes a defective recruitment onto centrosomes and satellites. The OFD KIAA0753/OFIP mutant loses its capacity to interact with FOR20 and OFD1, which may be the molecular basis of the defect. We also show that KIAA0753/OFIP has microtubule-stabilizing activity. OFD1 and FOR20 are known to regulate the integrity of the centriole distal end, confirming that this structural element is a target of importance for pathogenic mutations in ciliopathies.


Asunto(s)
Centriolos/metabolismo , Centrosoma/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Síndromes Orofaciodigitales/metabolismo , Proteínas/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Centriolos/ultraestructura , Centrosoma/ultraestructura , Cilios/genética , Cilios/metabolismo , Cilios/patología , Secuencia Conservada , Femenino , Expresión Génica , Heterocigoto , Humanos , Recién Nacido , Proteínas Asociadas a Microtúbulos/genética , Datos de Secuencia Molecular , Mutación , Síndromes Orofaciodigitales/genética , Síndromes Orofaciodigitales/patología , Unión Proteica , Proteínas/genética , Alineación de Secuencia
16.
PLoS One ; 9(8): e104302, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25144364

RESUMEN

BACKGROUND: Breast cancer stem cells (BCSCs) have been recognized as playing a major role in various aspects of breast cancer biology. To identify specific biomarkers of BCSCs, we have performed comparative proteomics of BCSC-enriched and mature cancer cell populations from the human breast cancer cell line (BCL), BrCA-MZ-01. METHODS: ALDEFLUOR assay was used to sort BCSC-enriched (ALDH+) and mature cancer (ALDH-) cell populations. Total proteins were extracted from both fractions and subjected to 2-Dimensional Difference In-Gel Electrophoresis (2-D DIGE). Differentially-expressed spots were excised and proteins were gel-extracted, digested and identified using MALDI-TOF MS. RESULTS: 2-D DIGE identified poly(ADP-ribose) polymerase 1 (PARP1) as overexpressed in ALDH+ cells from BrCA-MZ-01. This observation was confirmed by western blot and extended to four additional human BCLs. ALDH+ cells from BRCA1-mutated HCC1937, which had the highest level of PARP1 overexpression, displayed resistance to olaparib, a specific PARP1 inhibitor. CONCLUSION: An unbiased proteomic approach identified PARP1 as upregulated in ALDH+, BCSC-enriched cells from various human BCLs, which may contribute to clinical resistance to PARP inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/enzimología , Células Madre Neoplásicas/enzimología , Ftalazinas/farmacología , Piperazinas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Electroforesis en Gel Bidimensional , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
17.
J Proteome Res ; 13(5): 2478-94, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24654937

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a very aggressive malignancy characterized by an excessive resistance to all known anticancer therapies, a still largely elusive phenomenon. To identify original mechanisms, we have explored the role of post-translational modifications (PTMs) mediated by members of the ubiquitin family. Although alterations of these pathways have been reported in different cancers, no methodical search for these kinds of anomalies has been performed so far. Therefore, we studied the ubiquitin-, Nedd8-, and SUMO1-specific proteomes of a pancreatic cancer cell line (MiaPaCa-2) and identified changes induced by gemcitabine, the standard PDAC's chemotherapeutic drug. These PTMs profiles contained both known major substrates of all three modifiers as well as original ones. Gemcitabine treatment altered the PTM profile of proteins involved in various biological functions, some known cancer associated genes, many potentially cancer-associated genes, and several cancer-signaling networks, including canonical and noncanonical WNT and PI3K/Akt/MTOR pathways. Some of these altered PTMs formed groups of functionally and physically associated proteins. Importantly, we could validate the gemcitabine-induced PTMs variations of relevant candidates and we could demonstrate the biological significance of such altered PTMs by studying in detail the sumoylation of SNIP1, one of these new targets.


Asunto(s)
Antineoplásicos/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas/metabolismo , Transducción de Señal/efectos de los fármacos , Ubiquitina/metabolismo , Western Blotting , Línea Celular Tumoral , Cromatografía Liquida , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Microscopía Fluorescente , Proteína NEDD8 , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al ARN , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Espectrometría de Masas en Tándem , Ubiquitina/genética , Ubiquitinas/genética , Ubiquitinas/metabolismo , Proteína Wnt1/metabolismo , Gemcitabina
18.
Mol Biol Cell ; 25(5): 658-68, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24403606

RESUMEN

Formins constitute a large family of proteins that regulate the dynamics and organization of both the actin and microtubule cytoskeletons. Previously we showed that the formin mDia1 helps tether microtubules at the cell cortex, acting downstream of the ErbB2 receptor tyrosine kinase. Here we further study the contributions of mDia1 and its two most closely related formins, mDia2 and mDia3, to cortical microtubule capture and ErbB2-dependent breast carcinoma cell migration. We find that depletion of each of these three formins strongly disrupts chemotaxis without significantly affecting actin-based structures. Further, all three formins are required for formation of cortical microtubules in a nonredundant manner, and formin proteins defective in actin polymerization remain active for microtubule capture. Using affinity purification and mass spectrometry analysis, we identify differential binding partners of the formin-homology domain 2 (FH2) of mDia1, mDia2, and mDia3, which may explain their nonredundant roles in microtubule capture. The FH2 domain of mDia1 specifically interacts with Rab6-interacting protein 2 (Rab6IP2). Further, mDia1 is required for cortical localization of Rab6IP2, and concomitant depletion of Rab6IP2 and IQGAP1 severely disrupts cortical capture of microtubules, demonstrating the coinvolvement of mDia1, IQGAP1, and Rab6IP2 in microtubule tethering at the leading edge.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas Portadoras/fisiología , Movimiento Celular , Microtúbulos/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Quimiotaxis , Forminas , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Hierro no Heme/metabolismo , Estructura Terciaria de Proteína , Conejos , Receptor ErbB-2/metabolismo , Transducción de Señal
19.
BMC Genomics ; 13: 614, 2012 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-23148599

RESUMEN

BACKGROUND: Antibody responses against Anopheles salivary proteins can indicate individual exposure to bites of malaria vectors. The extent to which these salivary proteins are species-specific is not entirely resolved. Thus, a better knowledge of the diversity among salivary protein repertoires from various malaria vector species is necessary to select relevant genus-, subgenus- and/or species-specific salivary antigens. Such antigens could be used for quantitative (mosquito density) and qualitative (mosquito species) immunological evaluation of malaria vectors/host contact. In this study, salivary gland protein repertoires (sialomes) from several Anopheles species were compared using in silico analysis and proteomics. The antigenic diversity of salivary gland proteins among different Anopheles species was also examined. RESULTS: In silico analysis of secreted salivary gland protein sequences retrieved from an NCBInr database of six Anopheles species belonging to the Cellia subgenus (An. gambiae, An. arabiensis, An. stephensi and An. funestus) and Nyssorhynchus subgenus (An. albimanus and An. darlingi) displayed a higher degree of similarity compared to salivary proteins from closely related Anopheles species. Additionally, computational hierarchical clustering allowed identification of genus-, subgenus- and species-specific salivary proteins. Proteomic and immunoblot analyses performed on salivary gland extracts from four Anopheles species (An. gambiae, An. arabiensis, An. stephensi and An. albimanus) indicated that heterogeneity of the salivary proteome and antigenic proteins was lower among closely related anopheline species and increased with phylogenetic distance. CONCLUSION: This is the first report on the diversity of the salivary protein repertoire among species from the Anopheles genus at the protein level. This work demonstrates that a molecular diversity is exhibited among salivary proteins from closely related species despite their common pharmacological activities. The involvement of these proteins as antigenic candidates for genus-, subgenus- or species-specific immunological evaluation of individual exposure to Anopheles bites is discussed.


Asunto(s)
Anopheles/genética , Insectos Vectores/genética , Filogenia , Proteoma/genética , Glándulas Salivales/metabolismo , Animales , Anopheles/metabolismo , Secuencia de Bases , Análisis por Conglomerados , Biología Computacional , Electroforesis en Gel de Poliacrilamida , Immunoblotting , Insectos Vectores/metabolismo , Espectrometría de Masas , Proteómica , Alineación de Secuencia , Especificidad de la Especie
20.
PLoS One ; 7(9): e46213, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029439

RESUMEN

BACKGROUND: Mutations in the Planar Cell Polarity (PCP) core gene Vangl2 cause the most severe neural tube defects (NTD) in mice and humans. Genetic studies show that the Vangl2 gene genetically interacts with a close homologue Vangl1. How precisely Vangl2 and Vangl1 proteins interact and crosstalk has remained a difficult issue to address, with the main obstacle being the accurate discrimination of the two proteins, which share close sequence homology. Experimental evidence previously presented has been sparse and addressed with ectopically expressed proteins or with antibodies unable to biochemically discriminate Vangl1 from Vangl2, therefore giving rise to unclear results. METHODOLOGY AND MAIN FINDINGS: A highly specific monoclonal anti-Vangl2 antibody was generated and rigorously tested on both recombinant and extracted Vangl2 using surface plasmon resonance (SPR) analysis, western blot, and immunoprecipitation experiments. This antibody efficiently affinity-purified Vangl2 from cell lysates and allowed the unambiguous identification of endogenous Vangl2 by proteomic analysis. Vangl1 was also present in Vangl2 immunoprecipitates, establishing the first biochemical evidence for the existence of Vangl2/Vangl1 heterodimers at an endogenous level. Epitope-tagged Vangl2 and Vangl1 confirmed that both proteins interact and colocalize at the plasma membrane. The Vangl2 antibody is able to acutely assess differential expression levels of Vangl2 protein in culture cell lines, as corroborated with gene expression analysis. We characterised Vangl2 expression in the cochlea of homozygous and heterozygous Lp mutant mice bearing a point mutation within the C-terminal Vangl2 region that leads to profound PCP defects. Our antibody could detect much lower levels of Vangl2(Lp) protein in mutant mice compared to the wild type mice. CONCLUSION: Our results provide an in-depth biochemical characterisation of the interaction observed between Vangl paralogues.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Proteínas Portadoras/química , Proteínas de la Membrana/química , Proteínas del Tejido Nervioso/química , Defectos del Tubo Neural/genética , Mutación Puntual , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Membrana Celular/genética , Membrana Celular/metabolismo , Polaridad Celular/genética , Expresión Génica , Heterocigoto , Homocigoto , Humanos , Inmunoprecipitación , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Defectos del Tubo Neural/diagnóstico , Unión Proteica , Multimerización de Proteína , Proteómica , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...