Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.931
Filtrar
Más filtros

Intervalo de año de publicación
1.
Immunity ; 36(6): 959-73, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22705106

RESUMEN

RIG-I and MDA5 detect viral RNA in the cytoplasm and activate signaling cascades leading to the production of type-I interferons. RIG-I is activated through sequential binding of viral RNA and unanchored lysine-63 (K63) polyubiquitin chains, but how polyubiquitin activates RIG-I and whether MDA5 is activated through a similar mechanism remain unresolved. Here, we showed that the CARD domains of MDA5 bound to K63 polyubiquitin and that this binding was essential for MDA5 to activate the transcription factor IRF3. Mutations of conserved residues in MDA5 and RIG-I that disrupt their ubiquitin binding also abrogated their ability to activate IRF3. Polyubiquitin binding induced the formation of a large complex consisting of four RIG-I and four ubiquitin chains. This hetero-tetrameric complex was highly potent in activating the antiviral signaling cascades. These results suggest a unified mechanism of RIG-I and MDA5 activation and reveal a unique mechanism by which ubiquitin regulates cell signaling and immune response.


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Virus de la Encefalomiocarditis/fisiología , Poliubiquitina/metabolismo , Animales , Sistema Libre de Células , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/genética , Virus de la Encefalomiocarditis/genética , Fibroblastos/metabolismo , Fibroblastos/virología , Células HEK293/metabolismo , Células HEK293/virología , Humanos , Factor 3 Regulador del Interferón/metabolismo , Helicasa Inducida por Interferón IFIH1 , Interferón beta/biosíntesis , Interferón beta/genética , Ratones , Complejos Multiproteicos , Mutagénesis Sitio-Dirigida , Unión Proteica , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , ARN Viral/metabolismo , Receptores Inmunológicos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/fisiología , Transducción de Señal/fisiología , Relación Estructura-Actividad , Ubiquitinación
2.
Immunity ; 36(6): 933-46, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22749352

RESUMEN

The mitochondrial protein MAVS (also known as IPS-1, VISA, and CARDIF) interacts with RIG-I-like receptors (RLRs) to induce type I interferon (IFN-I). NLRX1 is a mitochondrial nucleotide-binding, leucine-rich repeats (NLR)-containing protein that attenuates MAVS-RLR signaling. Using Nlrx1(-/-) cells, we confirmed that NLRX1 attenuated IFN-I production, but additionally promoted autophagy during viral infection. This dual function of NLRX1 paralleled the previously described functions of the autophagy-related proteins Atg5-Atg12, but NLRX1 did not associate with Atg5-Atg12. High-throughput quantitative mass spectrometry and endogenous protein-protein interaction revealed an NLRX1-interacting partner, mitochondrial Tu translation elongation factor (TUFM). TUFM interacted with Atg5-Atg12 and Atg16L1 and has similar functions as NLRX1 by inhibiting RLR-induced IFN-I but promoting autophagy. In the absence of NLRX1, increased IFN-I and decreased autophagy provide an advantage for host defense against vesicular stomatitis virus. This study establishes a link between an NLR protein and the viral-induced autophagic machinery via an intermediary partner, TUFM.


Asunto(s)
Autofagia/fisiología , Interferón Tipo I/biosíntesis , Proteínas Mitocondriales/fisiología , Factor Tu de Elongación Peptídica/fisiología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Secuencia de Aminoácidos , Animales , Proteína 12 Relacionada con la Autofagia , Proteína 5 Relacionada con la Autofagia , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/fisiología , Citocinas/biosíntesis , Citocinas/genética , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/fisiología , Fibroblastos/metabolismo , Regulación de la Expresión Génica/inmunología , Células HEK293 , Humanos , Interferón Tipo I/genética , Macrófagos Peritoneales/citología , Macrófagos Peritoneales/inmunología , Ratones , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/fisiología , Proteínas Mitocondriales/química , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Datos de Secuencia Molecular , Complejos Multiproteicos/fisiología , Factor Tu de Elongación Peptídica/química , Mapeo de Interacción de Proteínas , Proteínas/fisiología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/fisiología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Organismos Libres de Patógenos Específicos , Vesiculovirus/fisiología
3.
Mol Cell ; 41(1): 33-45, 2011 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-21211721

RESUMEN

PARP-3 is a member of the ADP-ribosyl transferase superfamily of unknown function. We show that PARP-3 is stimulated by DNA double-strand breaks (DSBs) in vitro and functions in the same pathway as the poly (ADP-ribose)-binding protein APLF to accelerate chromosomal DNA DSB repair. We implicate PARP-3 in the accumulation of APLF at DSBs and demonstrate that APLF promotes the retention of XRCC4/DNA ligase IV complex in chromatin, suggesting that PARP-3 and APLF accelerate DNA ligation during nonhomologous end-joining (NHEJ). Consistent with this, we show that class switch recombination in Aplf(-/-) B cells is biased toward microhomology-mediated end-joining, a pathway that operates in the absence of XRCC4/DNA ligase IV, and that the requirement for PARP-3 and APLF for NHEJ is circumvented by overexpression of XRCC4/DNA ligase IV. These data identify molecular roles for PARP-3 and APLF in chromosomal DNA double-strand break repair reactions.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de Ciclo Celular/fisiología , Fosfoproteínas/fisiología , Poli(ADP-Ribosa) Polimerasas/fisiología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , ADN-(Sitio Apurínico o Apirimidínico) Liasa , Eliminación de Gen , Humanos , Ratones , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa , Proteínas Recombinantes de Fusión/fisiología
4.
EMBO J ; 31(2): 429-42, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22085926

RESUMEN

Wnts are required for cardiogenesis but the role of specific Wnts in cardiac repair remains unknown. In this report, we show that a dynamic Wnt1/ßcatenin injury response activates the epicardium and cardiac fibroblasts to promote cardiac repair. Acute ischaemic cardiac injury upregulates Wnt1 that is initially expressed in the epicardium and subsequently by cardiac fibroblasts in the region of injury. Following cardiac injury, the epicardium is activated organ-wide in a Wnt-dependent manner, expands, undergoes epithelial-mesenchymal transition (EMT) to generate cardiac fibroblasts, which localize in the subepicardial space. The injured regions in the heart are Wnt responsive as well and Wnt1 induces cardiac fibroblasts to proliferate and express pro-fibrotic genes. Disruption of downstream Wnt signalling in epicardial cells decreases epicardial expansion, EMT and leads to impaired cardiac function and ventricular dilatation after cardiac injury. Furthermore, disruption of Wnt/ßcatenin signalling in cardiac fibroblasts impairs wound healing and decreases cardiac performance as well. These findings reveal that a pro-fibrotic Wnt1/ßcatenin injury response is critically required for preserving cardiac function after acute ischaemic cardiac injury.


Asunto(s)
Fibroblastos/metabolismo , Corazón/fisiología , Infarto del Miocardio/patología , Pericardio/metabolismo , Regeneración/fisiología , Transducción de Señal/fisiología , Proteína Wnt1/fisiología , beta Catenina/fisiología , Animales , División Celular , Transición Epitelial-Mesenquimal , Fibrosis , Regulación de la Expresión Génica , Hibridación in Situ , Ratones , Ratones Transgénicos , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Pericardio/patología , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/fisiología , Regulación hacia Arriba , Proteína Wnt1/biosíntesis , Proteína Wnt1/genética , Cicatrización de Heridas/fisiología
5.
EMBO J ; 31(2): 417-28, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22036948

RESUMEN

Mobilization of intracellular Ca(2+) stores regulates a multitude of cellular functions, but the role of intracellular Ca(2+) release via the ryanodine receptor (RyR) in the brain remains incompletely understood. We found that nitric oxide (NO) directly activates RyRs, which induce Ca(2+) release from intracellular stores of central neurons, and thereby promote prolonged Ca(2+) signalling in the brain. Reversible S-nitrosylation of type 1 RyR (RyR1) triggers this Ca(2+) release. NO-induced Ca(2+) release (NICR) is evoked by type 1 NO synthase-dependent NO production during neural firing, and is essential for cerebellar synaptic plasticity. NO production has also been implicated in pathological conditions including ischaemic brain injury, and our results suggest that NICR is involved in NO-induced neuronal cell death. These findings suggest that NICR via RyR1 plays a regulatory role in the physiological and pathophysiological functions of the brain.


Asunto(s)
Señalización del Calcio/fisiología , Cerebelo/fisiología , Corteza Cerebral/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Óxido Nítrico/fisiología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Animales , Apoptosis/efectos de los fármacos , Cerebelo/citología , Corteza Cerebral/citología , Células HEK293 , Humanos , Técnicas In Vitro , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/fisiología , Técnicas de Placa-Clamp , Proteínas Recombinantes de Fusión/fisiología , Canal Liberador de Calcio Receptor de Rianodina/biosíntesis , Canal Liberador de Calcio Receptor de Rianodina/deficiencia , Canal Liberador de Calcio Receptor de Rianodina/genética
6.
EMBO J ; 31(2): 471-80, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22068052

RESUMEN

In addition to acting as a transcriptional cofactor for p53, ASPP1 has been shown to function in the cytoplasm to regulate the nuclear localization and activity of YAP/TAZ. We show here that the ability of ASPP1 to activate YAP results in the decreased expression of LATS2, which lowers the ability of p53 to induce p21, cell-cycle arrest and senescence. ASPP1 expression peaks in S-phase, and down-regulation of ASPP1 leads to a reduction in DNA synthesis and enhanced senescence in response to drugs that impede DNA replication. These activities of cytoplasmic ASPP1 in opposing p53-mediated p21 expression are in contrast to the role of nuclear ASPP1 in cooperating with p53 to induce the expression of apoptotic target genes, and may help to dampen p53 activity in normal cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas Reguladoras de la Apoptosis/fisiología , Senescencia Celular/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Regulación de la Expresión Génica/fisiología , Proteína p53 Supresora de Tumor/fisiología , Aciltransferasas , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Adenocarcinoma/patología , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Ciclo Celular , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Senescencia Celular/efectos de los fármacos , Neoplasias del Colon/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Replicación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/fisiología , Dactinomicina/farmacología , Doxorrubicina/farmacología , Humanos , Hidroxiurea/farmacología , Proteínas Nucleares/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/fisiología , Factores de Transcripción/fisiología , Transcripción Genética , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/fisiología
7.
EMBO J ; 30(16): 3274-84, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21743437

RESUMEN

Protein ubiquitylation is a key process in the regulation of many cellular processes. The balance between the activity of ubiquitin ligases and that of proteases controls the level of ubiquitylation. In response to extracellular stimuli, stress-activated protein kinases (SAPK) modulate gene expression to maximize cell survival. In yeast, the Hog1 SAPK has a key role in reprogramming the gene expression pattern required for cell survival upon osmostress. Here, we show that the Ubp3 ubiquitin protease is a target for the Hog1 SAPK to modulate gene expression. ubp3 mutant cells are defective in expression of osmoresponsive genes. Hog1 interacts with and phosphorylates Ubp3 at serine 695, which is essential to determine the extent of transcriptional activation in response to osmostress. Furthermore, Ubp3 is recruited to osmoresponsive genes to modulate transcriptional initiation as well as elongation. Therefore, Ubp3 activity responds to external stimuli and is required for transcriptional activation upon osmostress.


Asunto(s)
Endopeptidasas/fisiología , Regulación Fúngica de la Expresión Génica , Proteínas Quinasas Activadas por Mitógenos/fisiología , Procesamiento Proteico-Postraduccional , Proteínas de Saccharomyces cerevisiae/fisiología , Transcripción Genética , Activación Transcripcional , Endopeptidasas/biosíntesis , Endopeptidasas/genética , Eliminación de Gen , Sistema de Señalización de MAP Quinasas , Presión Osmótica/fisiología , Fosforilación , ARN Polimerasa II/metabolismo , ARN de Hongos/biosíntesis , ARN de Hongos/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/fisiología , Proteínas de Saccharomyces cerevisiae/biosíntesis , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Ubiquitinación
8.
EMBO J ; 30(16): 3309-21, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21743438

RESUMEN

Centromeres nucleate the formation of kinetochores and are vital for chromosome segregation during mitosis. The SNF2 family helicase PICH (Plk1-interacting checkpoint helicase) and the BLM (the Bloom's syndrome protein) helicase decorate ultrafine histone-negative DNA threads that link the segregating sister centromeres during anaphase. The functions of PICH and BLM at these threads are not understood, however. Here, we show that PICH binds to BLM and enables BLM localization to anaphase centromeric threads. PICH- or BLM-RNAi cells fail to resolve these threads in anaphase. The fragmented threads form centromeric-chromatin-containing micronuclei in daughter cells. Anaphase threads in PICH- and BLM-RNAi cells contain histones and centromere markers. Recombinant purified PICH has nucleosome remodelling activities in vitro. We propose that PICH and BLM unravel centromeric chromatin and keep anaphase DNA threads mostly free of nucleosomes, thus allowing these threads to span long distances between rapidly segregating centromeres without breakage and providing a spatiotemporal window for their resolution.


Asunto(s)
Centrómero/metabolismo , ADN Helicasas/fisiología , ADN/metabolismo , Nucleosomas/metabolismo , RecQ Helicasas/fisiología , Anafase , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Células HeLa , Histonas/metabolismo , Humanos , Micronúcleos con Defecto Cromosómico , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/fisiología
9.
EMBO J ; 30(16): 3397-415, 2011 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-21792176

RESUMEN

Hypoxia stabilizes the tumour suppressor p53, allowing it to function primarily as a transrepressor; however, the function of p53 during hypoxia remains unclear. In this study, we showed that p53 suppressed BNIP3 expression by directly binding to the p53-response element motif and recruiting corepressor mSin3a to the BNIP3 promoter. The DNA-binding site of p53 must remain intact for the protein to suppress the BNIP3 promoter. In addition, taking advantage of zebrafish as an in vivo model, we confirmed that zebrafish nip3a, a homologous gene of mammalian BNIP3, was indeed induced by hypoxia and p53 mutation/knockdown enhanced nip3a expression under hypoxia resulted in cell death enhancement in p53 mutant embryos. Furthermore, p53 protected against hypoxia-induced cell death mediated by p53 suppression of BNIP3 as illustrated by p53 knockdown/loss assays in both human cell lines and zebrafish model, which is in contrast to the traditional pro-apoptotic role of p53. Our results suggest a novel function of p53 in hypoxia-induced cell death, leading to the development of new treatments for ischaemic heart disease and cerebral stroke.


Asunto(s)
Apoptosis/fisiología , Hipoxia de la Célula/fisiología , Proteínas de la Membrana/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteína p53 Supresora de Tumor/fisiología , Proteínas de Pez Cebra/fisiología , Secuencia de Aminoácidos , Animales , Autofagia , Sitios de Unión , Línea Celular , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/fisiología , Elementos de Respuesta , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Pez Cebra/embriología
10.
EMBO J ; 30(16): 3259-73, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21765395

RESUMEN

Fibroblast growth factor receptor 1 (FGFR1) has critical roles in cellular proliferation and differentiation during animal development and adult homeostasis. Here, we show that human Nedd4 (Nedd4-1), an E3 ubiquitin ligase comprised of a C2 domain, 4 WW domains, and a Hect domain, regulates endocytosis and signalling of FGFR1. Nedd4-1 binds directly to and ubiquitylates activated FGFR1, by interacting primarily via its WW3 domain with a novel non-canonical sequence (non-PY motif) on FGFR1. Deletion of this recognition motif (FGFR1-Δ6) abolishes Nedd4-1 binding and receptor ubiquitylation, and impairs endocytosis of activated receptor, as also observed upon Nedd4-1 knockdown. Accordingly, FGFR1-Δ6, or Nedd4-1 knockdown, exhibits sustained FGF-dependent receptor Tyr phosphorylation and downstream signalling (activation of FRS2α, Akt, Erk1/2, and PLCγ). Expression of FGFR1-Δ6 in human embryonic neural stem cells strongly promotes FGF2-dependent neuronal differentiation. Furthermore, expression of this FGFR1-Δ6 mutant in zebrafish embryos disrupts anterior neuronal patterning (head development), consistent with excessive FGFR1 signalling. These results identify Nedd4-1 as a key regulator of FGFR1 endocytosis and signalling during neuronal differentiation and embryonic development.


Asunto(s)
Endocitosis/fisiología , Complejos de Clasificación Endosomal Requeridos para el Transporte/fisiología , Procesamiento Proteico-Postraduccional , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Tipificación del Cuerpo/fisiología , Diferenciación Celular/fisiología , Sistema Nervioso Central/embriología , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Técnicas de Silenciamiento del Gen , Humanos , Datos de Secuencia Molecular , Ubiquitina-Proteína Ligasas Nedd4 , Neuronas/citología , Fragmentos de Péptidos/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Ratas , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/química , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Recombinantes de Fusión/fisiología , Transducción de Señal/fisiología , Especificidad de la Especie , Células Madre/citología , Especificidad por Sustrato , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Pez Cebra/embriología
11.
EMBO J ; 30(16): 3383-96, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21750527

RESUMEN

Genomic instability due to telomere dysfunction and defective repair of DNA double-strand breaks (DSBs) is an underlying cause of ageing-related diseases. 53BP1 is a key factor in DNA DSBs repair and its deficiency is associated with genomic instability and cancer progression. Here, we uncover a novel pathway regulating the stability of 53BP1. We demonstrate an unprecedented role for the cysteine protease Cathepsin L (CTSL) in the degradation of 53BP1. Overexpression of CTSL in wild-type fibroblasts leads to decreased 53BP1 protein levels and changes in its cellular distribution, resulting in defective repair of DNA DSBs. Importantly, we show that the defects in DNA repair associated with 53BP1 deficiency upon loss of A-type lamins are due to upregulation of CTSL. Furthermore, we demonstrate that treatment with vitamin D stabilizes 53BP1 and promotes DNA DSBs repair via inhibition of CTSL, providing an as yet unsuspected link between vitamin D action and DNA repair. Given that CTSL upregulation is a hallmark of cancer and progeria, regulation of this pathway could be of great therapeutic significance for these diseases.


Asunto(s)
Catepsina L/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Lamina Tipo A/fisiología , Vitamina D/fisiología , Animales , Calcitriol/farmacología , Catepsina L/antagonistas & inhibidores , Catepsina L/biosíntesis , Catepsina L/genética , Línea Celular , Precursores Enzimáticos/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Lamina Tipo A/deficiencia , Lamina Tipo A/genética , Leupeptinas/farmacología , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteínas Recombinantes de Fusión/fisiología , Especificidad de la Especie , Transfección , Proteína 1 de Unión al Supresor Tumoral P53
12.
EMBO J ; 30(16): 3337-52, 2011 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-21785410

RESUMEN

Spindle pole bodies (SPBs), like nuclear pore complexes, are embedded in the nuclear envelope (NE) at sites of fusion of the inner and outer nuclear membranes. A network of interacting proteins is required to insert a cytoplasmic SPB precursor into the NE. A central player of this network is Nbp1 that interacts with the conserved integral membrane protein Ndc1. Here, we establish that Nbp1 is a monotopic membrane protein that is essential for SPB insertion at the inner face of the NE. In vitro and in vivo studies identified an N-terminal amphipathic α-helix of Nbp1 as a membrane-binding element, with crucial functions in SPB duplication. The karyopherin Kap123 binds to a nuclear localization sequence next to this amphipathic α-helix and prevents unspecific tethering of Nbp1 to membranes. After transport into the nucleus, Nbp1 binds to the inner nuclear membrane. These data define the targeting pathway of a SPB component and suggest that the amphipathic α-helix of Nbp1 is important for SPB insertion into the NE from within the nucleus.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Proteínas de Ciclo Celular/fisiología , Proteínas del Citoesqueleto/fisiología , Membrana Nuclear/metabolismo , Proteínas Nucleares/fisiología , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomyces cerevisiae/metabolismo , Huso Acromático/metabolismo , beta Carioferinas/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/genética , Liposomas/metabolismo , Fusión de Membrana , Datos de Secuencia Molecular , Señales de Localización Nuclear , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosfatidilcolinas/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/fisiología , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Eliminación de Secuencia , Relación Estructura-Actividad , beta Carioferinas/genética
13.
Blood ; 121(1): 178-87, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23152544

RESUMEN

The origin of aberrant DNA methylation in cancer remains largely unknown. In the present study, we elucidated the DNA methylome in primary acute promyelocytic leukemia (APL) and the role of promyelocytic leukemia-retinoic acid receptor α (PML-RARα) in establishing these patterns. Cells from APL patients showed increased genome-wide DNA methylation with higher variability than healthy CD34(+) cells, promyelocytes, and remission BM cells. A core set of differentially methylated regions in APL was identified. Age at diagnosis, Sanz score, and Flt3-mutation status characterized methylation subtypes. Transcription factor-binding sites (eg, the c-myc-binding sites) were associated with low methylation. However, SUZ12- and REST-binding sites identified in embryonic stem cells were preferentially DNA hypermethylated in APL cells. Unexpectedly, PML-RARα-binding sites were also protected from aberrant DNA methylation in APL cells. Consistent with this, myeloid cells from preleukemic PML-RARα knock-in mice did not show altered DNA methylation and the expression of PML-RARα in hematopoietic progenitor cells prevented differentiation without affecting DNA methylation. Treatment of APL blasts with all-trans retinoic acid also did not result in immediate DNA methylation changes. The results of the present study suggest that aberrant DNA methylation is associated with leukemia phenotype but is not required for PML-RARα-mediated initiation of leukemogenesis.


Asunto(s)
Metilación de ADN , ADN de Neoplasias/genética , Regulación Leucémica de la Expresión Génica , Leucemia Promielocítica Aguda/genética , Factores de Transcripción/metabolismo , Animales , Transformación Celular Neoplásica/genética , Cromosomas Humanos/ultraestructura , Islas de CpG , ADN de Neoplasias/metabolismo , Progresión de la Enfermedad , Técnicas de Sustitución del Gen , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/metabolismo , Proteínas de Fusión Oncogénica/fisiología , Fenotipo , Complejo Represivo Polycomb 2/metabolismo , Preleucemia/genética , Proteínas Recombinantes de Fusión/fisiología , Proteínas Represoras/metabolismo , Tretinoina/uso terapéutico
14.
Blood ; 121(1): 72-84, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23160469

RESUMEN

Wiskott Aldrich syndrome (WAS), an X-linked immunodeficiency, results from loss-of-function mutations in the human hematopoietic cytoskeletal regulator gene WAS. Many missense mutations in the Ena Vasp homology1 (EVH1) domain preserve low-level WAS protein (WASp) expression and confer a milder clinical phenotype. Although disrupted binding to WASp-interacting protein (WIP) leads to enhanced WASp degradation in vivo, the intrinsic function of EVH1-mutated WASp is poorly understood. In the present study, we show that, despite mediating enhanced actin polymerization compared with wild-type WASp in vitro, EVH1 missense mutated proteins did not support full biologic function in cells, even when levels were restored by forced overexpression. Podosome assembly was aberrant and associated with dysregulated lamellipodia formation and impaired persistence of migration. At sites of residual podosome-associated actin polymerization, localization of EVH1-mutated proteins was preserved even after deletion of the entire domain, implying that WIP-WASp complex formation is not absolutely required for WASp localization. However, retention of mutant proteins in podosomes was significantly impaired and associated with reduced levels of WASp tyrosine phosphorylation. Our results indicate that the EVH1 domain is important not only for WASp stability, but also for intrinsic biologic activity in vivo.


Asunto(s)
Células Dendríticas/patología , Mutación Missense , Proteína del Síndrome de Wiskott-Aldrich/genética , Actinas/metabolismo , Animales , Biopolímeros , Proteínas Portadoras/metabolismo , Movimiento Celular , Células Cultivadas , Proteínas del Citoesqueleto , Células Dendríticas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fosforilación , Polimerizacion , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Estructura Terciaria de Proteína , Seudópodos/patología , Proteínas Recombinantes de Fusión/fisiología , Eliminación de Secuencia , Organismos Libres de Patógenos Específicos , Proteína del Síndrome de Wiskott-Aldrich/química , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/fisiología
15.
Blood ; 121(3): 447-58, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23169777

RESUMEN

To search for genes that promote hematopoietic development from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs), we overexpressed several known hematopoietic regulator genes in hESC/iPSC-derived CD34(+)CD43(-) endothelial cells (ECs) enriched in hemogenic endothelium (HE). Among the genes tested, only Sox17, a gene encoding a transcription factor of the SOX family, promoted cell growth and supported expansion of CD34(+)CD43(+)CD45(-/low) cells expressing the HE marker VE-cadherin. SOX17 was expressed at high levels in CD34(+)CD43(-) ECs compared with low levels in CD34(+)CD43(+)CD45(-) pre-hematopoietic progenitor cells (pre-HPCs) and CD34(+)CD43(+)CD45(+) HPCs. Sox17-overexpressing cells formed semiadherent cell aggregates and generated few hematopoietic progenies. However, they retained hemogenic potential and gave rise to hematopoietic progenies on inactivation of Sox17. Global gene-expression analyses revealed that the CD34(+)CD43(+)CD45(-/low) cells expanded on overexpression of Sox17 are HE-like cells developmentally placed between ECs and pre-HPCs. Sox17 overexpression also reprogrammed both pre-HPCs and HPCs into HE-like cells. Genome-wide mapping of Sox17-binding sites revealed that Sox17 activates the transcription of key regulator genes for vasculogenesis, hematopoiesis, and erythrocyte differentiation directly. Depletion of SOX17 in CD34(+)CD43(-) ECs severely compromised their hemogenic activity. These findings suggest that SOX17 plays a key role in priming hemogenic potential in ECs, thereby regulating hematopoietic development from hESCs/iPSCs.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/fisiología , Animales , Diferenciación Celular/fisiología , División Celular/fisiología , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/citología , Células Endoteliales/fisiología , Sangre Fetal/citología , Fibroblastos/citología , Hematopoyesis/genética , Humanos , Lentivirus/genética , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Transducción Genética/métodos
16.
Blood ; 121(1): 159-69, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23100311

RESUMEN

Recently, we showed that increased miR-181a expression was associated with improved outcomes in cytogenetically normal acute myeloid leukemia (CN-AML). Interestingly, miR-181a expression was increased in CN-AML patients harboring CEBPA mutations, which are usually biallelic and associate with better prognosis. CEBPA encodes the C/EBPα transcription factor. We demonstrate here that the presence of N-terminal CEBPA mutations and miR-181a expression are linked. Indeed, the truncated C/EBPα-p30 isoform, which is produced from the N-terminal mutant CEBPA gene or from the differential translation of wild-type CEBPA mRNA and is commonly believed to have no transactivation activity, binds to the miR-181a-1 promoter and up-regulates the microRNA expression. Furthermore, we show that lenalidomide, a drug approved for myelodysplastic syndromes and multiple myeloma, enhances translation of the C/EBPα-p30 isoform, resulting in higher miR-181a levels. In xenograft mouse models, ectopic miR-181a expression inhibits tumor growth. Similarly, lenalidomide exhibits antitumorigenic activity paralleled by increased miR-181a expression. This regulatory pathway may explain an increased sensitivity to apoptosis-inducing chemotherapy in subsets of AML patients. Altogether, our data provide a potential explanation for the improved clinical outcomes observed in CEBPA-mutated CN-AML patients, and suggest that lenalidomide treatment enhancing the C/EBPα-p30 protein levels and in turn miR-181a may sensitize AML blasts to chemotherapy.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/fisiología , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Factores Inmunológicos/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , MicroARNs/biosíntesis , Proteínas de Neoplasias/biosíntesis , ARN Neoplásico/biosíntesis , Talidomida/análogos & derivados , Adulto , Animales , Antimetabolitos Antineoplásicos/farmacología , Proteínas Potenciadoras de Unión a CCAAT/biosíntesis , Proteínas Potenciadoras de Unión a CCAAT/genética , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Línea Celular Tumoral/trasplante , Citarabina/farmacología , Mutación del Sistema de Lectura , Humanos , Factores Inmunológicos/uso terapéutico , Células K562 , Lenalidomida , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/genética , Proteínas de Neoplasias/genética , Mutación Puntual , Regiones Promotoras Genéticas/genética , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , ARN Neoplásico/genética , Proteínas Recombinantes de Fusión/fisiología , Talidomida/farmacología , Talidomida/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Circ Res ; 112(4): 664-74, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23283722

RESUMEN

RATIONALE: Myocardial diastolic stiffness and cardiomyocyte passive force (F(passive)) depend in part on titin isoform composition and phosphorylation. Ca(2+)/calmodulin-dependent protein kinase-II (CaMKII) phosphorylates ion channels, Ca(2+)-handling proteins, and chromatin-modifying enzymes in the heart, but has not been known to target titin. OBJECTIVE: To elucidate whether CaMKII phosphorylates titin and modulates F(passive) in normal and failing myocardium. METHODS AND RESULTS: Titin phosphorylation was assessed in CaMKIIδ/γ double-knockout (DKO) mouse, transgenic CaMKIIδC-overexpressing mouse, and human hearts, by Pro-Q-Diamond/Sypro-Ruby staining, autoradiography, and immunoblotting using phosphoserine-specific titin-antibodies. CaMKII-dependent site-specific titin phosphorylation was quantified in vivo by mass spectrometry using stable isotope labeling by amino acids in cell culture mouse heart mixed with wild-type (WT) or DKO heart. F(passive) of single permeabilized cardiomyocytes was recorded before and after CaMKII-administration. All-titin phosphorylation was reduced by >50% in DKO but increased by up to ≈100% in transgenic versus WT hearts. Conserved CaMKII-dependent phosphosites were identified within the PEVK-domain of titin by quantitative mass spectrometry and confirmed in recombinant human PEVK-fragments. CaMKII also phosphorylated the cardiac titin N2B-unique sequence. Phosphorylation at specific PEVK/titin N2B-unique sequence sites was decreased in DKO and amplified in transgenic versus WT hearts. F(passive) was elevated in DKO and reduced in transgenic compared with WT cardiomyocytes. CaMKII-administration lowered F(passive) of WT and DKO cardiomyocytes, an effect blunted by titin antibody pretreatment. Human end-stage failing hearts revealed higher CaMKII expression/activity and phosphorylation at PEVK/titin N2B-unique sequence sites than nonfailing donor hearts. CONCLUSIONS: CaMKII phosphorylates the titin springs at conserved serines/threonines, thereby lowering F(passive). Deranged CaMKII-dependent titin phosphorylation occurs in heart failure and contributes to altered diastolic stress.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Insuficiencia Cardíaca/enzimología , Proteínas Musculares/metabolismo , Proteínas Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Fenómenos Biomecánicos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/deficiencia , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Adaptabilidad , Conectina , Diástole/fisiología , Insuficiencia Cardíaca/fisiopatología , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/fisiología , Fosforilación , Fosfoserina/metabolismo , Fosfotreonina/metabolismo , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/fisiología
18.
Circ Res ; 113(1): 40-51, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23603512

RESUMEN

RATIONALE: Endothelial microRNA-126 (miR-126) modulates vascular development and angiogenesis. However, its role in the regulation of smooth muscle cell (SMC) function is unknown. OBJECTIVE: To elucidate the role of miR-126 secreted by endothelial cells (ECs) in regulating SMC turnover in vitro and in vivo, as well as the effects of shear stress on the regulation. METHODS AND RESULTS: Coculture of SMCs with ECs or treatment of SMCs with conditioned media from static EC monoculture (EC-CM) increased SMC miR-126 level and SMC turnover; these effects were abolished by inhibition of endothelial miR-126 and by the application of laminar shear stress to ECs. SMC miR-126 did not increase when treated with EC-CM from ECs subjected to inhibition of miR biogenesis, or with CM from sheared ECs. Depletion of extracellular/secreted vesicles in EC-CM did not affect the increase of SMC miR-126 by EC-CM. Biotinylated miR-126 or FLAG (DYKDDDDK epitope)-tagged Argonaute2 transfected into ECs was detected in the cocultured or EC-CM-treated SMCs, indicating a direct EC-to-SMC transmission of miR-126 and Argonaute2. Endothelial miR-126 represses forkhead box O3, B-cell lymphoma 2, and insulin receptor substrate 1 mRNAs in the cocultured SMCs, suggesting the functional roles of the transmitted miR-126. Systemic depletion of miR-126 in mice inhibited neointimal lesion formation of carotid arteries induced by cessation of blood flow. Administration of EC-CM or miR-126 mitigated the inhibitory effect. CONCLUSIONS: Endothelial miR-126 acts as a key intercellular mediator to increase SMC turnover, and its release is reduced by atheroprotective laminar shear stress.


Asunto(s)
Células Endoteliales/metabolismo , Regulación de la Expresión Génica , MicroARNs/fisiología , Miocitos del Músculo Liso/citología , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/fisiología , Arteria Carótida Común/patología , Técnicas de Cultivo de Célula/instrumentación , Línea Celular , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Silenciador del Gen , Genes bcl-2 , Hemorreología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas Sustrato del Receptor de Insulina/biosíntesis , Proteínas Sustrato del Receptor de Insulina/genética , Ligadura , Ratones , MicroARNs/genética , MicroARNs/uso terapéutico , Músculo Liso Vascular/citología , Neointima , Comunicación Paracrina , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Recombinantes de Fusión/fisiología , Arterias Umbilicales/citología
19.
Circ Res ; 113(5): 571-87, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23948583

RESUMEN

Recent advances in the burgeoning field of genome engineering are accelerating the realization of personalized therapeutics for cardiovascular disease. In the postgenomic era, sequence-specific gene-editing tools enable the functional analysis of genetic alterations implicated in disease. In partnership with high-throughput model systems, efficient gene manipulation provides an increasingly powerful toolkit to study phenotypes associated with patient-specific genetic defects. Herein, this review emphasizes the latest developments in genome engineering and how applications within the field are transforming our understanding of personalized medicine with an emphasis on cardiovascular diseases.


Asunto(s)
Proteínas Bacterianas/fisiología , Enfermedades Cardiovasculares/genética , Desoxirribonucleasas de Localización Especificada Tipo II/fisiología , Desoxirribonucleasas/fisiología , Ingeniería Genética/métodos , Genómica , Células Madre Pluripotentes Inducidas/citología , Mutagénesis Sitio-Dirigida/métodos , Medicina de Precisión/tendencias , Xanthomonas/enzimología , Animales , Proteínas Bacterianas/genética , Sitios de Unión , Enfermedades Cardiovasculares/terapia , Diferenciación Celular , Células Cultivadas/citología , Células Cultivadas/trasplante , ADN/genética , ADN/metabolismo , Desoxirribonucleasas/genética , Desoxirribonucleasas de Localización Especificada Tipo II/genética , Predicción , Genes Reporteros , Ingeniería Genética/tendencias , Humanos , Células Madre Pluripotentes Inducidas/trasplante , Modelos Cardiovasculares , Modelos Genéticos , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/fisiología , Reparación del ADN por Recombinación , Especificidad por Sustrato , Xanthomonas axonopodis/enzimología , Pez Cebra/genética
20.
Circ Res ; 112(4): 651-63, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23271793

RESUMEN

RATIONALE: Oxidation of cysteine residues in class II histone deacetylases (HDACs), including HDAC4, causes nuclear exit, thereby inducing cardiac hypertrophy. The cellular source of reactive oxygen species responsible for oxidation of HDAC4 remains unknown. OBJECTIVE: We investigated whether nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4), a major nicotinamide adenine dinucleotide phosphate oxidase, mediates cysteine oxidation of HDAC4. METHODS AND RESULTS: Phenylephrine (100 µmol/L), an α1 adrenergic agonist, induced upregulation of Nox4 (1.5-fold; P<0.05) within 5 minutes, accompanied by increases in O(2)(-) (3.5-fold; P<0.01) from the nuclear membrane and nuclear exit of HDAC4 in cardiomyocytes. Knockdown of Nox4, but not Nox2, attenuated O(2)(-) production in the nucleus and prevented phenylephrine-induced oxidation and nuclear exit of HDAC4. After continuous infusion of phenylephrine (20 mg/kg per day) for 14 days, wild-type and cardiac-specific Nox4 knockout mice exhibited similar aortic pressures. Left ventricular weight/tibial length (5.7±0.2 versus 6.4±0.2 mg/mm; P<0.05) and cardiomyocytes cross-sectional area (223±13 versus 258±12 µm(2); P<0.05) were significantly smaller in cardiac-specific Nox4 knockout than in wild-type mice. Nuclear O(2)(-)production in the heart was significantly lower in cardiac-specific Nox4 knockout than in wild-type mice (4116±314 versus 7057±1710 relative light unit; P<0.05), and cysteine oxidation of HDAC4 was decreased. HDAC4 oxidation and cardiac hypertrophy were also attenuated in cardiac-specific Nox4 knockout mice 2 weeks after transverse aortic constriction. CONCLUSIONS: Nox4 plays an essential role in mediating cysteine oxidation and nuclear exit of HDAC4, thereby mediating cardiac hypertrophy in response to phenylephrine and pressure overload.


Asunto(s)
Cardiomegalia/metabolismo , Histona Desacetilasas/metabolismo , Miocitos Cardíacos/efectos de los fármacos , NADPH Oxidasas/fisiología , Animales , Estenosis de la Válvula Aórtica/metabolismo , Cardiomegalia/etiología , Cardiomegalia/genética , Núcleo Celular/metabolismo , Tamaño de la Célula , Cisteína/metabolismo , Activación Enzimática , Inducción Enzimática/efectos de los fármacos , Masculino , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , NADPH Oxidasa 2 , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/deficiencia , NADPH Oxidasas/genética , Membrana Nuclear/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Fenilefrina/farmacología , Transporte de Proteínas/fisiología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA