Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
JCI Insight ; 6(1)2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33427209

RESUMEN

RNA-binding proteins (RBPs) are essential factors required for the physiological function of neurons, muscle, and other tissue types. In keeping with this, a growing body of genetic, clinical, and pathological evidence indicates that RBP dysfunction and/or gene mutation leads to neurodegeneration and myopathy. Here, we summarize the current understanding of matrin 3 (MATR3), a poorly understood RBP implicated not only in ALS and frontotemporal dementia but also in distal myopathy. We begin by reviewing MATR3's functions, its regulation, and how it may be involved in both sporadic and familial neuromuscular disease. We also discuss insights gleaned from cellular and animal models of MATR3 pathogenesis, the links between MATR3 and other disease-associated RBPs, and the mechanisms underlying RBP-mediated disorders.


Asunto(s)
Enfermedades Neuromusculares/genética , Enfermedades Neuromusculares/fisiopatología , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Modelos Animales de Enfermedad , Miopatías Distales/genética , Miopatías Distales/fisiopatología , Demencia Frontotemporal/genética , Demencia Frontotemporal/fisiopatología , Regulación de la Expresión Génica , Humanos , Ratones Noqueados , Mutación , Proteínas Asociadas a Matriz Nuclear/química , Proteínas Asociadas a Matriz Nuclear/deficiencia , Proteínas de Unión al ARN/química
2.
Nat Cell Biol ; 21(10): 1273-1285, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31548606

RESUMEN

Chromosome translocation is a major cause of the onset and progression of diverse types of cancers. However, the mechanisms underlying this process remain poorly understood. Here, we identified a non-homologous end-joining protein, IFFO1, which structurally forms a heterotetramer with XRCC4. IFFO1 is recruited to the sites of DNA damage by XRCC4 and promotes the repair of DNA double-strand breaks in a parallel pathway with XLF. Interestingly, IFFO1 interacts with lamin A/C, forming an interior nucleoskeleton. Inactivating IFFO1 or its interaction with XRCC4 or lamin A/C leads to increases in both the mobility of broken ends and the frequency of chromosome translocation. Importantly, the destruction of this nucleoskeleton accounts for the elevated frequency of chromosome translocation in many types of cancer cells. Our results reveal that the lamin A/C-IFFO1-constituted nucleoskeleton prevents chromosome translocation by immobilizing broken DNA ends during tumorigenesis.


Asunto(s)
Carcinogénesis/genética , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Lamina Tipo A/metabolismo , Translocación Genética , Animales , Carcinoma/genética , Cromosomas Humanos , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Células HEK293 , Humanos , Proteínas de Filamentos Intermediarios/genética , Ratones , Matriz Nuclear/metabolismo , Proteínas Asociadas a Matriz Nuclear/química , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas Asociadas a Matriz Nuclear/fisiología
3.
Sci Rep ; 9(1): 13530, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31537859

RESUMEN

Several cellular processes depend on networks of proteins assembled at specific sites near the plasma membrane. Scaffold proteins assemble these networks by recruiting relevant molecules. The scaffold protein ERC1/ELKS and its partners promote cell migration and invasion, and assemble into dynamic networks at the protruding edge of cells. Here by electron microscopy and single molecule analysis we identify ERC1 as an extended flexible dimer. We found that ERC1 scaffolds form cytoplasmic condensates with a behavior that is consistent with liquid phases that are modulated by a predicted disordered region of ERC1. These condensates specifically host partners of a network relevant to cell motility, including liprin-α1, which was unnecessary for the formation of condensates, but influenced their dynamic behavior. Phase separation at specific sites of the cell periphery may represent an elegant mechanism to control the assembly and turnover of dynamic scaffolds needed for the spatial localization and processing of molecules.


Asunto(s)
Movimiento Celular/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células COS , Línea Celular Tumoral , Membrana Celular/metabolismo , Chlorocebus aethiops , Citoplasma/metabolismo , Humanos , Proteínas del Tejido Nervioso/fisiología , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas Asociadas a Matriz Nuclear/fisiología , Proteínas de Unión al GTP rab/fisiología
5.
Vet Res ; 50(1): 22, 2019 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-30894203

RESUMEN

Nuclear localization of paramyxovirus proteins is crucial for virus life cycle, including the regulation of viral replication and the evasion of host immunity. We previously showed that a recombinant Newcastle disease virus (NDV) with nuclear localization signal mutation in the matrix (M) protein results in a pathotype change and attenuates viral pathogenicity in chickens. However, little is known about the nuclear localization functions of NDV M protein. In this study, the potential functions of the M protein in the nucleus were investigated. We first demonstrate that nuclear localization of the M protein could not only promote the cytopathogenicity of NDV but also increase viral RNA synthesis and transcription efficiency in DF-1 cells. Using microarray analysis, we found that nuclear localization of the M protein might inhibit host cell transcription, represented by numerous up-regulating genes associated with transcriptional repressor activity and down-regulating genes associated with transcriptional activator activity. The role of representative up-regulated gene prospero homeobox 1 (PROX1) and down-regulated gene aryl hydrocarbon receptor (AHR) in the replication of NDV was then evaluated. The results show that siRNA-mediated knockdown of PROX1 or AHR significantly reduced or increased the viral RNA synthesis and viral replication, respectively, demonstrating the important roles of the expression changes of these genes in NDV replication. Together, our findings demonstrate for the first time that nuclear localization of NDV M protein promotes virus replication by affecting viral RNA synthesis and transcription and inhibiting host cell transcription, improving our understanding of the molecular mechanism of NDV replication and pathogenesis.


Asunto(s)
Fibroblastos/virología , Proteínas Asociadas a Matriz Nuclear/fisiología , Transporte de Proteínas/fisiología , ARN Viral/metabolismo , Transcripción Genética , Replicación Viral/fisiología , Animales , Línea Celular , Pollos , Regulación Viral de la Expresión Génica/fisiología , Virus de la Enfermedad de Newcastle , ARN Viral/genética
6.
EMBO J ; 38(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30787184

RESUMEN

Deubiquitination of NLRP3 has been suggested to contribute to inflammasome activation, but the roles and molecular mechanisms are still unclear. We here demonstrate that ABRO1, a subunit of the BRISC deubiquitinase complex, is necessary for optimal NLRP3-ASC complex formation, ASC oligomerization, caspase-1 activation, and IL-1ß and IL-18 production upon treatment with NLRP3 ligands after the priming step, indicating that efficient NLRP3 activation requires ABRO1. Moreover, we report that ABRO1 deficiency results in a remarkable attenuation in the syndrome severity of NLRP3-associated inflammatory diseases, including MSU- and Alum-induced peritonitis and LPS-induced sepsis in mice. Mechanistic studies reveal that LPS priming induces ABRO1 binding to NLRP3 in an S194 phosphorylation-dependent manner, subsequently recruiting the BRISC to remove K63-linked ubiquitin chains of NLRP3 upon stimulation with activators. Furthermore, deficiency of BRCC3, the catalytically active component of BRISC, displays similar phenotypes to ABRO1 knockout mice. Our findings reveal an ABRO1-mediated regulatory signaling system that controls activation of the NLRP3 inflammasome and provide novel potential targets for treating NLRP3-associated inflammatory diseases.


Asunto(s)
Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Proteínas Asociadas a Matriz Nuclear/fisiología , Peritonitis/etiología , Proteasas Ubiquitina-Específicas/fisiología , Ubiquitinación , Ubiquitinas/metabolismo , Animales , Enzimas Desubicuitinizantes/fisiología , Femenino , Inflamasomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peritonitis/metabolismo , Peritonitis/patología , Fosforilación , Proteolisis , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
7.
Neurobiol Learn Mem ; 159: 36-45, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30790622

RESUMEN

microRNA-dependent post-transcriptional control represents an important gene-regulatory layer in post-mitotic neuronal development and synaptic plasticity. We recently identified the brain-enriched miR-138 as a negative regulator of dendritic spine morphogenesis in rat hippocampal neurons. A potential involvement of miR-138 in cognition is further supported by a recent GWAS study on memory performance in a cohort of aged (>60 years) individuals. The expression of miR-138, which is encoded in two independent genomic loci (miR-138-1 and -2), is subject to both cell-type and developmental stage-specific regulation, the underlying molecular mechanisms however are poorly understood. Here, we show that miR-138-2 is the primary source of mature miR-138 in developing rat hippocampal neurons. Furthermore, we obtained evidence for the regulation of miR-138-2 biogenesis at the level of primary miRNA processing. Using biochemical pull-down assays, we identified the nuclear matrix protein Matrin-3 as pri/pre-miR-138 interacting protein and mapped the interaction to the pri/pre-miR-138-2 loop region. Matrin-3 loss-of-function experiments in HEK293 cells and primary neurons together with protein localization studies suggest an inhibitory function of Matrin-3 in nuclear pri-miR-138-2 processing. Together, our experiments unravel a new mechanism of miR-138 regulation in neurons, with important implications for miR-138 regulation during neuronal development, synaptic plasticity and memory-related processes.


Asunto(s)
Hipocampo/metabolismo , MicroARNs/metabolismo , Plasticidad Neuronal/fisiología , Proteínas Asociadas a Matriz Nuclear/fisiología , Proteínas de Unión al ARN/fisiología , Animales , Embrión de Mamíferos , Células HEK293 , Humanos , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas
8.
Cell ; 176(4): 716-728.e18, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30712871

RESUMEN

Sensory axons degenerate following separation from their cell body, but partial injury to peripheral nerves may leave the integrity of damaged axons preserved. We show that an endogenous ligand for the natural killer (NK) cell receptor NKG2D, Retinoic Acid Early 1 (RAE1), is re-expressed in adult dorsal root ganglion neurons following peripheral nerve injury, triggering selective degeneration of injured axons. Infiltration of cytotoxic NK cells into the sciatic nerve by extravasation occurs within 3 days following crush injury. Using a combination of genetic cell ablation and cytokine-antibody complex stimulation, we show that NK cell function correlates with loss of sensation due to degeneration of injured afferents and reduced incidence of post-injury hypersensitivity. This neuro-immune mechanism of selective NK cell-mediated degeneration of damaged but intact sensory axons complements Wallerian degeneration and suggests the therapeutic potential of modulating NK cell function to resolve painful neuropathy through the clearance of partially damaged nerves.


Asunto(s)
Células Asesinas Naturales/fisiología , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Animales , Axones , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Células Asesinas Naturales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Regeneración Nerviosa , Neuronas/citología , Neuronas Aferentes/inmunología , Neuronas Aferentes/metabolismo , Proteínas Asociadas a Matriz Nuclear/fisiología , Proteínas de Transporte Nucleocitoplasmático/fisiología , Dolor , Traumatismos de los Nervios Periféricos/inmunología , Enfermedades del Sistema Nervioso Periférico , Nervio Ciático , Células Receptoras Sensoriales/metabolismo
9.
Nucleic Acids Res ; 47(6): 2703-2715, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30812030

RESUMEN

P53-binding protein 1 (53BP1) mediates DNA repair pathway choice and promotes checkpoint activation. Chromatin marks induced by DNA double-strand breaks and recognized by 53BP1 enable focal accumulation of this multifunctional repair factor at damaged chromatin. Here, we unveil an additional level of regulation of 53BP1 outside repair foci. 53BP1 movements are constrained throughout the nucleoplasm and increase in response to DNA damage. 53BP1 interacts with the structural protein NuMA, which controls 53BP1 diffusion. This interaction, and colocalization between the two proteins in vitro and in breast tissues, is reduced after DNA damage. In cell lines and breast carcinoma NuMA prevents 53BP1 accumulation at DNA breaks, and high NuMA expression predicts better patient outcomes. Manipulating NuMA expression alters PARP inhibitor sensitivity of BRCA1-null cells, end-joining activity, and immunoglobulin class switching that rely on 53BP1. We propose a mechanism involving the sequestration of 53BP1 by NuMA in the absence of DNA damage. Such a mechanism may have evolved to disable repair functions and may be a decisive factor for tumor responses to genotoxic treatments.


Asunto(s)
Antígenos Nucleares/fisiología , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Proteínas Asociadas a Matriz Nuclear/fisiología , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Reparación del ADN por Unión de Extremidades/genética , Regulación hacia Abajo , Femenino , Células HEK293 , Humanos , Unión Proteica
10.
Cell ; 175(2): 488-501.e22, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30270045

RESUMEN

Detection of viruses by innate immune sensors induces protective antiviral immunity. The viral DNA sensor cyclic GMP-AMP synthase (cGAS) is necessary for detection of HIV by human dendritic cells and macrophages. However, synthesis of HIV DNA during infection is not sufficient for immune activation. The capsid protein, which associates with viral DNA, has a pivotal role in enabling cGAS-mediated immune activation. We now find that NONO is an essential sensor of the HIV capsid in the nucleus. NONO protein directly binds capsid with higher affinity for weakly pathogenic HIV-2 than highly pathogenic HIV-1. Upon infection, NONO is essential for cGAS activation by HIV and cGAS association with HIV DNA in the nucleus. NONO recognizes a conserved region in HIV capsid with limited tolerance for escape mutations. Detection of nuclear viral capsid by NONO to promote DNA sensing by cGAS reveals an innate strategy to achieve distinction of viruses from self in the nucleus.


Asunto(s)
Proteínas de la Cápside/inmunología , Proteínas Asociadas a Matriz Nuclear/inmunología , Proteínas Asociadas a Matriz Nuclear/fisiología , Factores de Transcripción de Octámeros/inmunología , Factores de Transcripción de Octámeros/fisiología , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/fisiología , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/fisiología , Núcleo Celular/metabolismo , ADN Viral/genética , ADN Viral/inmunología , Proteínas de Unión al ADN , Células Dendríticas/inmunología , Infecciones por VIH/inmunología , VIH-1/genética , VIH-1/inmunología , VIH-2/genética , VIH-2/inmunología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata/inmunología , Macrófagos/inmunología , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/fisiología , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/inmunología
11.
Endocrinology ; 159(11): 3615-3630, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30204866

RESUMEN

Among their pleiotropic functions, scaffold proteins are required for the accurate coordination of signaling pathways. It has only been within the past 10 years that their roles in glucose homeostasis and metabolism have emerged. It is well appreciated that changes in the expression or function of signaling effectors, such as receptors or kinases, can influence the development of chronic diseases such as diabetes and obesity. However, little is known regarding whether scaffolds have similar roles in the pathogenesis of metabolic diseases. In general, scaffolds are often underappreciated in the context of metabolism or metabolic diseases. In the present review, we discuss various scaffold proteins and their involvement in signaling pathways related to metabolism and metabolic diseases. The aims of the present review were to highlight the importance of scaffold proteins and to raise awareness of their physiological contributions. A thorough understanding of how scaffolds influence metabolism could aid in the discovery of novel therapeutic approaches to treat chronic conditions, such as diabetes, obesity, and cardiovascular disease, for which the incidence of all continue to increase at alarming rates.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Diabetes Mellitus/metabolismo , Resistencia a la Insulina/fisiología , Secreción de Insulina/fisiología , Proteínas Asociadas a Matriz Nuclear/metabolismo , Obesidad/metabolismo , Transducción de Señal/fisiología , Adiposidad/fisiología , Animales , Apoptosis/fisiología , Glucemia/metabolismo , Metabolismo Energético , Gluconeogénesis/fisiología , Homeostasis , Humanos , Enfermedades Metabólicas/metabolismo , Proteínas Asociadas a Matriz Nuclear/fisiología
12.
Genes Dev ; 31(14): 1469-1482, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28860160

RESUMEN

Protection of the stalled replication fork is crucial for responding to replication stress and minimizing its impact on chromosome instability, thus preventing diseases, including cancer. We found a new component, Abro1, in the protection of stalled replication fork integrity. Abro1 deficiency results in increased chromosome instability, and Abro1-null mice are tumor-prone. We show that Abro1 protects stalled replication fork stability by inhibiting DNA2 nuclease/WRN helicase-mediated degradation of stalled forks. Depletion of RAD51 prevents the DNA2/WRN-dependent degradation of stalled forks in Abro1-deficient cells. This mechanism is distinct from the BRCA2-dependent fork protection pathway, in which stable RAD51 filament formation prevents MRE11-dependent degradation of the newly synthesized DNA at stalled forks. Thus, our data reveal a new aspect of regulated protection of stalled replication forks that involves Abro1.


Asunto(s)
Replicación del ADN , Inestabilidad Genómica , Proteínas Asociadas a Matriz Nuclear/fisiología , Proteasas Ubiquitina-Específicas/fisiología , Animales , Proteína BRCA2/genética , Línea Celular , Células Cultivadas , ADN/biosíntesis , ADN Helicasas/fisiología , Endodesoxirribonucleasas/fisiología , Proteína Homóloga de MRE11/fisiología , Ratones Noqueados , Enzimas Multifuncionales/fisiología , Neoplasias Experimentales/genética , Proteínas Asociadas a Matriz Nuclear/genética , Recombinasa Rad51/genética , Estrés Fisiológico , Proteasas Ubiquitina-Específicas/genética
13.
Yakugaku Zasshi ; 136(3): 485-90, 2016.
Artículo en Japonés | MEDLINE | ID: mdl-26935091

RESUMEN

Cl(-)-permeable channels and transporters expressed on the cell membranes of various mammalian cell types play pivotal roles in the transport of electrolytes and water, pH regulation, cell volume and membrane excitability, and are therefore expected to be useful molecular targets for drug discovery. Both TMEM16A (a possible candidate for Ca(2+)-regulated Cl(-) channels recently identified) and cystic fibrosis transmembrane conductance regulator (CFTR) (or cAMP-regulated Cl(-) channels) have been known to be involved in Cl(-) secretion and reabsorption in the rat salivary gland. Crosstalk between two types of regulatory pathways through these two types of channels has also been described. Previously, we demonstrated that CLCA, a Ca(2+)-activated Cl(-) channel modulator, was involved in Cl(-) absorption in rat salivary ducts. In addition to Ca(2+), basal NF-κB activity in a mouse keratinocyte line was shown to be involved in the transcriptional regulation of CLCA. Conversely, a truncated isoform of CLCA was found in undifferentiated epithelial cells present in the rat epidermal basal layers. Under regulation by Ca(2+) and PKC, the surface expression of ß1-integrin and cell adhesion were decreased in the CLCA-overexpressing cells. Knockdown of this isoform elevated the expression of ß1-integrin in rat epidermis in vivo. These results indicate that the specific differentiation-dependent localization of CLCA, and transcriptional regulation through Ca(2+), are likely to affect ion permeability and the adhesive potential of epithelial cells. In summary, these types of Cl(-) channels and their modulators may function in a coordinated manner in regulating the functions of epithelial cells under different physiological conditions.


Asunto(s)
Diferenciación Celular/genética , Canales de Cloruro/fisiología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Hierro/metabolismo , Animales , Anoctamina-1 , Calcio/fisiología , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Cloruros/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Humanos , Ratones , FN-kappa B/fisiología , Proteínas de Neoplasias/fisiología , Proteínas Asociadas a Matriz Nuclear/fisiología , Ratas , Glándulas Salivales/metabolismo , Transcripción Genética
14.
Neurobiol Aging ; 37: 209.e17-209.e21, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26493020

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder characterized by an extensive loss of motor neurons in the primary motor cortex, brainstem, and spinal cord. Genetic studies report a high heritability of ALS. Recently, whole-exome sequencing analysis of familial ALS (FALS) patients allowed the identification of missense variations within the MATR3 gene. MATR3 was previously associated to distal myopathy 2 and encodes for a nuclear matrix and DNA/RNA binding protein that has been shown to interact with TDP43 in an RNA-dependent manner. Here, we assessed the MATR3 mutation frequency in French-Canadian ALS and control individuals (nFALS = 83, sporadic ALS [nSALS] = 164, and ncontrols = 162) and showed that MATR3 mutations were found in 0%, 1.8%, and 0% of FALS, SALS, and controls, respectively. Interestingly, among the mutations identified in SALS, the splicing mutation c.48+1G>T was found to result in the insertion of 24 amino acids in MATR3 protein. These findings further support the role of MATR3 in ALS, and more studies are needed to shed more light on MATR3 proteinopathy.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Estudios de Asociación Genética , Mutación , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas de Unión al ARN/genética , Secuencia de Bases , Canadá , Proteínas de Unión al ADN/genética , Exoma/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Datos de Secuencia Molecular , Proteínas Asociadas a Matriz Nuclear/fisiología , ARN/metabolismo , Proteínas de Unión al ARN/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Población Blanca
15.
Oncogene ; 35(15): 2003-10, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-26165835

RESUMEN

Genotoxic stresses lead to centrosome amplification, a frequently-observed feature in cancer that may contribute to genome instability and to tumour cell invasion. Here we have explored how the centrosome controls DNA damage responses. For most of the cell cycle, centrosomes consist of two centrioles embedded in the proteinaceous pericentriolar material (PCM). Recent data indicate that the PCM is not an amorphous assembly of proteins, but actually a highly organised scaffold around the centrioles. The large coiled-coil protein, pericentrin, participates in PCM assembly and has been implicated in the control of DNA damage responses (DDRs) through its interactions with checkpoint kinase 1 (CHK1) and microcephalin (MCPH1). CHK1 is required for DNA damage-induced centrosome amplification, whereas MCPH1 deficiency greatly increases the amplification seen after DNA damage. We found that the PCM showed a marked expansion in volume and a noticeable change in higher-order organisation after ionising radiation treatment. PCM expansion was dependent on CHK1 kinase activity and was potentiated by MCPH1 deficiency. Furthermore, pericentrin deficiency or mutation of a separase cleavage site blocked DNA damage-induced PCM expansion. The extent of nuclear CHK1 activation after DNA damage reflected the level of PCM expansion, with a reduction in pericentrin-deficient or separase cleavage site mutant-expressing cells, and an increase in MCPH1-deficient cells that was suppressed by the loss of pericentrin. Deletion of the nuclear export signal of CHK1 led to its hyperphosphorylation after irradiation and reduced centrosome amplification. Deletion of the nuclear localisation signal led to low CHK1 activation and low centrosome amplification. From these data, we propose a feedback loop from the PCM to the nuclear DDR in which CHK1 regulates pericentrin-dependent PCM expansion to control its own activation.


Asunto(s)
Antígenos/fisiología , Proteínas de Ciclo Celular/fisiología , Centrosoma/fisiología , Daño del ADN , Proteínas del Tejido Nervioso/fisiología , Proteínas Asociadas a Matriz Nuclear/fisiología , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Animales , Antígenos/genética , Sitios de Unión , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Centriolos/metabolismo , Centriolos/ultraestructura , Centrosoma/efectos de la radiación , Centrosoma/ultraestructura , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Pollos , Activación Enzimática , Retroalimentación Fisiológica , Genes Reporteros , Mutación , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Proteínas Asociadas a Matriz Nuclear/genética , Fosforilación , Proteínas Recombinantes de Fusión/metabolismo , Separasa/metabolismo
16.
Oncogene ; 35(5): 567-76, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-25893301

RESUMEN

The main risk factor for skin cancer is ultraviolet (UV) exposure, which causes DNA damage. Cells respond to UV-induced DNA damage by activating the intra-S-phase checkpoint, which prevents replication fork collapse, late origin firing and stabilizes fragile sites. Recently, the 54-kDa multifunctional protein NONO was found to be involved in the non-homologous end-joining DNA repair process and in poly ADP-ribose polymerase 1 activation. Interestingly, NONO is mutated in several tumour types and emerged as a crucial factor underlying both melanoma development and progression. Therefore, we set out to evaluate whether NONO could be involved in the DNA-damage response to UV radiations. We generated NONO-silenced HeLa cell clones and found that lack of NONO decreased cell growth rate. Then, we challenged NONO-silenced cells with exposure to UV radiations and found that NONO-silenced cells, compared with control cells, continued to synthesize DNA, failed to block new origin firing and impaired CHK1S345 phosphorylation showing a defective checkpoint activation. Consistently, NONO is present at the sites of UV-induced DNA damage where it localizes to RAD9 foci. To position NONO in the DNA-damage response cascade, we analysed the loading onto chromatin of various intra-S-phase checkpoint mediators and found that NONO favours the loading of topoisomerase II-binding protein 1 acting upstream of the ATM and Rad3-related kinase activity. Strikingly, re-expression of NONO, through an sh-resistant mRNA, rescued CHK1S345 phosphorylation in NONO-silenced cells. Interestingly, NONO silencing affected cell response to UV radiations also in a melanoma cell line. Overall, our data uncover a new role for NONO in mediating the cellular response to UV-induced DNA damage.


Asunto(s)
Daño del ADN , Proteínas Asociadas a Matriz Nuclear/fisiología , Factores de Transcripción de Octámeros/fisiología , Proteínas de Unión al ARN/fisiología , Puntos de Control de la Fase S del Ciclo Celular/fisiología , Puntos de Control de la Fase S del Ciclo Celular/efectos de la radiación , ADN/metabolismo , Reparación del ADN , Proteínas de Unión al ADN , Células HeLa , Humanos , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Factores de Transcripción de Octámeros/genética , Factores de Transcripción de Octámeros/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Puntos de Control de la Fase S del Ciclo Celular/genética , Transfección , Rayos Ultravioleta
17.
Mol Cell ; 59(6): 970-83, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26344097

RESUMEN

BRCC36 is a Zn(2+)-dependent deubiquitinating enzyme (DUB) that hydrolyzes lysine-63-linked ubiquitin chains as part of distinct macromolecular complexes that participate in either interferon signaling or DNA-damage recognition. The MPN(+) domain protein BRCC36 associates with pseudo DUB MPN(-) proteins KIAA0157 or Abraxas, which are essential for BRCC36 enzymatic activity. To understand the basis for BRCC36 regulation, we have solved the structure of an active BRCC36-KIAA0157 heterodimer and an inactive BRCC36 homodimer. Structural and functional characterizations show how BRCC36 is switched to an active conformation by contacts with KIAA0157. Higher-order association of BRCC36 and KIAA0157 into a dimer of heterodimers (super dimers) was required for DUB activity and interaction with targeting proteins SHMT2 and RAP80. These data provide an explanation of how an inactive pseudo DUB allosterically activates a cognate DUB partner and implicates super dimerization as a new regulatory mechanism underlying BRCC36 DUB activity, subcellular localization, and biological function.


Asunto(s)
Hormigas/enzimología , Proteínas de Insectos/química , Proteínas Asociadas a Matriz Nuclear/química , Proteasas Ubiquitina-Específicas/química , Animales , Dominio Catalítico , Cristalografía por Rayos X , Enzimas Desubicuitinizantes , Células HEK293 , Células HeLa , Humanos , Proteínas de Insectos/fisiología , Cinética , Proteínas de la Membrana/química , Modelos Moleculares , Proteínas Asociadas a Matriz Nuclear/fisiología , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Proteasas Ubiquitina-Específicas/fisiología
18.
Nucleic Acids Res ; 43(7): 3605-13, 2015 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-25800734

RESUMEN

Early steps of gene expression are a composite of promoter recognition, promoter activation, RNA synthesis and RNA processing, and it is known that SUMOylation, a post-translational modification, is involved in transcription regulation. We previously found that SUMO-1 marks chromatin at the proximal promoter regions of some of the most active housekeeping genes during interphase in human cells, but the SUMOylated targets on the chromatin remained unclear. In this study, we found that SUMO-1 marks the promoters of ribosomal protein genes via modification of the Scaffold Associated Factor B (SAFB) protein, and the SUMOylated SAFB stimulated both the binding of RNA polymerase to promoters and pre-mRNA splicing. Depletion of SAFB decreased RNA polymerase II binding to promoters and nuclear processing of the mRNA, though mRNA stability was not affected. This study reveals an unexpected role of SUMO-1 and SAFB in the stimulatory coupling of promoter binding, transcription initiation and RNA processing.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/fisiología , Proteínas Asociadas a Matriz Nuclear/fisiología , Regiones Promotoras Genéticas , Empalme del ARN , Receptores de Estrógenos/fisiología , Proteínas Ribosómicas/genética , Proteína SUMO-1/metabolismo , Transcripción Genética/fisiología , Regulación hacia Abajo , Células HeLa , Humanos , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo
19.
J Cell Biol ; 206(2): 245-56, 2014 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-25023517

RESUMEN

The spindle is a dynamic self-assembling machine that coordinates mitosis. The spindle's function depends on its ability to organize microtubules into poles and maintain pole structure despite mechanical challenges and component turnover. Although we know that dynein and NuMA mediate pole formation, our understanding of the forces dynamically maintaining poles is limited: we do not know where and how quickly they act or their strength and structural impact. Using laser ablation to cut spindle microtubules, we identify a force that rapidly and robustly pulls severed microtubules and chromosomes poleward, overpowering opposing forces and repairing spindle architecture. Molecular imaging and biophysical analysis suggest that transport is powered by dynein pulling on minus ends of severed microtubules. NuMA and dynein/dynactin are specifically enriched at new minus ends within seconds, reanchoring minus ends to the spindle and delivering them to poles. This force on minus ends represents a newly uncovered chromosome transport mechanism that is independent of plus end forces at kinetochores and is well suited to robustly maintain spindle mechanical integrity.


Asunto(s)
Cromosomas/metabolismo , Microtúbulos/fisiología , Huso Acromático/fisiología , Animales , Transporte Biológico , Línea Celular , Complejo Dinactina , Dineínas/metabolismo , Dineínas/fisiología , Cinetocoros , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/fisiología , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas Asociadas a Matriz Nuclear/fisiología , Potoroidae , Huso Acromático/metabolismo , Huso Acromático/ultraestructura
20.
Semin Cell Dev Biol ; 34: 140-5, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24972323

RESUMEN

The angle of cell division is critical in at least two contexts. It can determine cell fate, as it does in developing neural tissue. It can also dictate tissue architecture, as it does in many epithelia. One way to ensure the correct angle of cell division is through controlled orientation of the spindle at metaphase. What happens when that control is lost? Ongoing work suggests that the consequence of metaphase spindle misorientation may be significant, but multiple mechanisms exist to protect the cell and the tissue. We speculate that one such mechanism involves a recently identified anaphase activity for two of the key players at metaphase: NuMA (Mud, LIN-5) and dynein.


Asunto(s)
Huso Acromático/metabolismo , Animales , Antígenos Nucleares/fisiología , Carcinogénesis/patología , Proteínas de Ciclo Celular , Sistema Nervioso Central/patología , Humanos , Mitosis , Neoplasias/patología , Proteínas Asociadas a Matriz Nuclear/fisiología , Huso Acromático/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...