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.
Nucleic Acids Res ; 48(21): e122, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33053171

RESUMEN

Protein-protein interactions are essential to ensure timely and precise recruitment of chromatin remodellers and repair factors to DNA damage sites. Conventional analyses of protein-protein interactions at a population level may mask the complexity of interaction dynamics, highlighting the need for a method that enables quantification of DNA damage-dependent interactions at a single-cell level. To this end, we integrated a pulsed UV laser on a confocal fluorescence lifetime imaging (FLIM) microscope to induce localized DNA damage. To quantify protein-protein interactions in live cells, we measured Förster resonance energy transfer (FRET) between mEGFP- and mCherry-tagged proteins, based on the fluorescence lifetime reduction of the mEGFP donor protein. The UV-FLIM-FRET system offers a unique combination of real-time and single-cell quantification of DNA damage-dependent interactions, and can distinguish between direct protein-protein interactions, as opposed to those mediated by chromatin proximity. Using the UV-FLIM-FRET system, we show the dynamic changes in the interaction between poly(ADP-ribose) polymerase 1, amplified in liver cancer 1, X-ray repair cross-complementing protein 1 and tripartite motif containing 33 after DNA damage. This new set-up complements the toolset for studying DNA damage response by providing single-cell quantitative and dynamic information about protein-protein interactions at DNA damage sites.


Asunto(s)
Osteoblastos/efectos de la radiación , Poli(ADP-Ribosa) Polimerasa-1/genética , Mapeo de Interacción de Proteínas/métodos , Factores de Transcripción/genética , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/genética , Línea Celular Tumoral , Cromatina/química , Cromatina/metabolismo , Cromatina/efectos de la radiación , Daño del ADN , Transferencia Resonante de Energía de Fluorescencia , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Rayos Láser , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Imagen Óptica , Osteoblastos/citología , Osteoblastos/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Unión Proteica , Transducción de Señal , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Rayos Ultravioleta , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/metabolismo , Proteína Fluorescente Roja
2.
Cells ; 9(2)2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32085595

RESUMEN

LAP2-Emerin-MAN1 (LEM) domain-containing proteins represent an abundant group of inner nuclear membrane proteins involved in diverse nuclear functions, but their functional redundancies remain unclear. Here, using the biotinylation-dependent proximity approach, we report proteome-wide comparative interactome analysis of the two structurally related LEM proteins MAN1 (LEMD3) and LEM2 (LEMD2), and the more distantly related emerin (EMD). While over 60% of the relatively small group of MAN1 and emerin interactors were also found in the LEM2 interactome, the latter included a large number of candidates (>85%) unique for LEM2. The interacting partners unique for emerin support and provide further insight into the previously reported role of emerin in centrosome positioning, and the MAN1-specific interactors suggest a role of MAN1 in ribonucleoprotein complex assembly. Interestingly, the LEM2-specific interactome contained several proteins of the nucleotide excision repair pathway. Accordingly, LEM2-depleted cells, but not MAN1- and emerin-depleted cells, showed impaired proliferation following ultraviolet-C (UV-C) irradiation and prolonged accumulation of γH2AX, similar to cells deficient in the nucleotide excision repair protein DNA damage-binding protein 1 (DDB1). These findings indicate impaired DNA damage repair in LEM2-depleted cells. Overall, this interactome study identifies new potential interaction partners of emerin, MAN1 and particularly LEM2, and describes a novel potential involvement of LEM2 in nucleotide excision repair at the nuclear periphery.


Asunto(s)
Reparación del ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Proliferación Celular/efectos de la radiación , Centrosoma/metabolismo , Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Histonas/metabolismo , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Dominios Proteicos , Transfección , Rayos Ultravioleta
3.
Nucleus ; 9(1): 474-491, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30205747

RESUMEN

Fluorescence microscopy in combination with the induction of localized DNA damage using focused light beams has played a major role in the study of protein recruitment kinetics to DNA damage sites in recent years. Currently published methods are dedicated to the study of single fluorophore/single protein kinetics. However, these methods may be limited when studying the relative recruitment dynamics between two or more proteins due to cell-to-cell variability in gene expression and recruitment kinetics, and are not suitable for comparative analysis of fast-recruiting proteins. To tackle these limitations, we have established a time-lapse fluorescence microscopy method based on simultaneous dual-channel acquisition following UV-A-induced local DNA damage coupled with a standardized image and recruitment analysis workflow. Simultaneous acquisition is achieved by spectrally splitting the emitted light into two light paths, which are simultaneously imaged on two halves of the same camera chip. To validate this method, we studied the recruitment of poly(ADP-ribose) polymerase 1 (PARP1), poly (ADP-ribose) glycohydrolase (PARG), proliferating cell nuclear antigen (PCNA) and the chromatin remodeler ALC1. In accordance with the published data based on single fluorophore imaging, simultaneous dual-channel imaging revealed that PARP1 regulates fast recruitment and dissociation of PARG and that in PARP1-depleted cells PARG and PCNA are recruited with comparable kinetics. This approach is particularly advantageous for analyzing the recruitment sequence of fast-recruiting proteins such as PARP1 and ALC1, and revealed that PARP1 is recruited faster than ALC1. Split-view imaging can be incorporated into any laser microirradiation-adapted microscopy setup together with a recruitment-dedicated image analysis package.


Asunto(s)
Daño del ADN , ADN Helicasas/análisis , Proteínas de Unión al ADN/análisis , Glicósido Hidrolasas/análisis , Rayos Láser , Imagen Óptica , Poli(ADP-Ribosa) Polimerasa-1/análisis , Antígeno Nuclear de Célula en Proliferación/análisis , Rayos Ultravioleta , Línea Celular Tumoral , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Glicósido Hidrolasas/metabolismo , Humanos , Cinética , Microscopía Fluorescente , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo
4.
Nucleic Acids Res ; 45(16): 9741-9759, 2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-28934471

RESUMEN

Poly(ADP-ribose) glycohydrolase (PARG) regulates cellular poly(ADP-ribose) (PAR) levels by rapidly cleaving glycosidic bonds between ADP-ribose units. PARG interacts with proliferating cell nuclear antigen (PCNA) and is strongly recruited to DNA damage sites in a PAR- and PCNA-dependent fashion. Here we identified PARG acetylation site K409 that is essential for its interaction with PCNA, its localization within replication foci and its recruitment to DNA damage sites. We found K409 to be part of a non-canonical PIP-box within the PARG disordered regulatory region. The previously identified putative N-terminal PIP-box does not bind PCNA directly but contributes to PARG localization within replication foci. X-ray structure and MD simulations reveal that the PARG non-canonical PIP-box binds PCNA in a manner similar to other canonical PIP-boxes and may represent a new type of PIP-box. While the binding of previously described PIP-boxes is based on hydrophobic interactions, PARG PIP-box binds PCNA via both stabilizing hydrophobic and fine-tuning electrostatic interactions. Our data explain the mechanism of PARG-PCNA interaction through a new PARG PIP-box that exhibits non-canonical sequence properties but a canonical mode of PCNA binding.


Asunto(s)
Glicósido Hidrolasas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Acetilación , Calorimetría/métodos , Cromatina/metabolismo , Cristalografía por Rayos X , Daño del ADN , Transferencia Resonante de Energía de Fluorescencia , Glicósido Hidrolasas/química , Glicósido Hidrolasas/genética , Células HeLa , Humanos , Inmunoprecipitación , Rayos Láser , Lisina/genética , Lisina/metabolismo , Simulación de Dinámica Molecular , Antígeno Nuclear de Célula en Proliferación/química , Conformación Proteica , Fase S/genética , Electricidad Estática
5.
J Cell Sci ; 125(Pt 4): 1048-57, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22399800

RESUMEN

The LEM domain (for lamina-associated polypeptide, emerin, MAN1 domain) defines a group of nuclear proteins that bind chromatin through interaction of the LEM motif with the conserved DNA crosslinking protein, barrier-to-autointegration factor (BAF). Here, we describe a LEM protein annotated in databases as 'Ankyrin repeat and LEM domain-containing protein 1' (Ankle1). We show that Ankle1 is conserved in metazoans and contains a unique C-terminal GIY-YIG motif that confers endonuclease activity in vitro and in vivo. In mammals, Ankle1 is predominantly expressed in hematopoietic tissues. Although most characterized LEM proteins are components of the inner nuclear membrane, ectopic Ankle1 shuttles between cytoplasm and nucleus. Ankle1 enriched in the nucleoplasm induces DNA cleavage and DNA damage response. This activity requires both the catalytic C-terminal GIY-YIG domain and the LEM motif, which binds chromatin via BAF. Hence, Ankle1 is an unusual LEM protein with a GIY-YIG-type endonuclease activity in higher eukaryotes.


Asunto(s)
División del ADN , Endonucleasas/química , Endonucleasas/metabolismo , Secuencias de Aminoácidos , Animales , Línea Celular , Núcleo Celular/metabolismo , Secuencia Conservada , Citoplasma/metabolismo , Daño del ADN , Endonucleasas/análisis , Endonucleasas/genética , Perfilación de la Expresión Génica , Sistema Hematopoyético/metabolismo , Humanos , Inmunoprecipitación , Especificidad de Órganos , Reacción en Cadena de la Polimerasa , Estructura Terciaria de Proteína , Transporte de Proteínas , Transducción de Señal
6.
Cell Mol Life Sci ; 68(9): 1593-610, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20922455

RESUMEN

Cell polarization is a fundamental process underpinning organismal development, and tissue homeostasis, which requires an orchestrated interplay of nuclear, cytoskeletal, and centrosomal structures. The underlying molecular mechanisms, however, still remain elusive. Here we report that kinesin-1/nesprin-2/SUN-domain macromolecular assemblies, spanning the entire nuclear envelope (NE), function in cell polarization by anchoring cytoskeletal structures to the nuclear lamina. Nesprin-2 forms complexes with the kinesin-1 motor protein apparatus by associating with and recruiting kinesin light chain 1 (KLC1) to the outer nuclear membrane. Similar to nesprin-2, KLC1 requires lamin A/C for proper NE localization. The depletion of nesprin-2 or KLC1, or the uncoupling of nesprin-2/SUN-domain protein associations impairs cell polarization during wounding and dislodges the centrosome from the NE. In addition nesprin-2 loss has profound effects on KLC1 levels, the cytoskeleton, and Golgi apparatus organization. Collectively these data show that NE-associated proteins are pivotal determinants of cell architecture and polarization.


Asunto(s)
Centrosoma/metabolismo , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Membrana Nuclear/metabolismo , Animales , Línea Celular , Polaridad Celular , Chlorocebus aethiops/metabolismo , Dineínas/metabolismo , Humanos , Cinesinas/metabolismo , Regiones de Fijación a la Matriz , Ratones , Proteínas del Tejido Nervioso/metabolismo
7.
Biochim Biophys Acta ; 1783(12): 2415-26, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18845190

RESUMEN

SUN-domain proteins form a novel and conserved family of inner nuclear membrane (INM) proteins, which establish physical connections between the nucleoplasm and the cytoskeleton. In the current study, we provide evidence that within the nuclear envelope (NE) Sun1 proteins form highly immobile oligomeric complexes in interphase cells. By performing inverse fluorescence recovery after photobleaching analysis, we demonstrate in vivo that both perinuclear and nucleoplasmic Sun1 segments are essential for maintenance of Sun1 immobility at the NE. Our data in particular underline the self-association properties of the C-terminal coiled-coil Sun1 segment, the ability of which to form dimers and tetramers is demonstrated. Furthermore, the Sun1 tertiary structure involves interchain disulfide bonds that might contribute to higher homo-oligomer formation, although the overall dynamics of the Sun1 C-terminus remains unaffected when the cysteins involved are mutated. While a major Sun1 pool colocalizes with nuclear pore complex proteins, a large fraction of the Sun1 protein assemblies colocalize with immunoreactive foci of Sun2, another SUN-domain paralogue at the NE. We demonstrate that the Sun1 coiled-coil domain permits these heterophilic associations with Sun2. Sun1 therefore provides a non-dynamic platform for the formation of different macromolecular assemblies at the INM. Our data support a model in which SUN-protein-containing multi-variate complexes may provide versatile outer nuclear membrane attachment sites for cytoskeletal filaments.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Poro Nuclear/fisiología , Secuencia de Aminoácidos , Western Blotting , Núcleo Celular/ultraestructura , Reactivos de Enlaces Cruzados , Disulfuros/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo , Técnica del Anticuerpo Fluorescente , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Células HeLa , Humanos , Técnicas para Inmunoenzimas , Proteínas Asociadas a Microtúbulos/genética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Membrana Nuclear/ultraestructura , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Técnicas del Sistema de Dos Híbridos
8.
FEBS J ; 274(6): 1362-73, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17489094

RESUMEN

Lamins are major structural components of the nuclear envelope in multicellular eukaryotes. Particularly A-type lamins are also located in the nucleoplasm, likely involving a specific binding partner, lamina-associated polypeptide 2alpha (LAP2alpha). LAP2alpha-lamins A/C complexes in the nucleoplasm have been implicated in the regulation of gene expression by various means. They bind chromatin proteins and chromatin modifying enzymes, and can thus participate in epigenetic control pathways. Furthermore, binding of lamins A/C complexes to specific transcription factors and repressors may directly affect their transcriptional activity. LAP2alpha-lamins A/C also regulate retinoblastoma protein and influence cell cycle progression and differentiation, which could have important implications for molecular mechanisms of laminopathic diseases, linked to lamins A/C mutations.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Laminas/fisiología , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/fisiología , Fosfoproteínas/fisiología , Proteína de Retinoblastoma/metabolismo , Transcripción Genética , Regulación de la Expresión Génica , Humanos , Laminas/metabolismo , Proteínas Nucleares/metabolismo , Nucleoplasminas , Fosfoproteínas/metabolismo , Unión Proteica
9.
Trends Cell Biol ; 17(4): 202-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17320395

RESUMEN

Barrier-to-autointegration factor (BAF) is an abundant, highly conserved, small and essential protein that binds to dsDNA, chromatin, nuclear lamina proteins, histones and various transcription factors. It was discovered as a cellular component of retrovirus pre-integration complex that inhibits their autointegration in vitro. BAF is also required for many cellular functions, including the higher-order organization of chromatin and the transcription of specific genes. Recent findings suggest further roles for BAF, including nuclear envelope assembly, regulating specific developmental processes and regulating retrovirus infectivity. At least some of these roles are controlled by phosphorylation of the BAF N-terminus by the vaccinia-related kinase. Here, we give an overview of recent advances in the field of BAF with special emphasis on evolution, interacting partners and functions.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas Nucleares/fisiología , Secuencia de Aminoácidos , Animales , Regulación de la Expresión Génica/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana , Datos de Secuencia Molecular , Membrana Nuclear/fisiología , Retroviridae/fisiología , Alineación de Secuencia , Integración Viral/fisiología
10.
J Cell Sci ; 120(Pt 5): 737-47, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17284516

RESUMEN

Lamina-associated polypeptide 2alpha (LAP2alpha) is a nuclear protein dynamically associating with chromatin during the cell cycle. In addition, LAP2alpha interacts with A-type lamins and retinoblastoma protein and regulates cell cycle progression via the E2F-Rb pathway. Using yeast two-hybrid analysis and three independent in vitro binding assays we identified a new LAP2alpha interaction partner of hitherto unknown functions, which we termed LINT-25. LINT-25 protein levels were upregulated during G1 phase in proliferating cells and upon cell cycle exit in quiescence, senescence and differentiation. Upon cell cycle exit LINT-25 accumulated in heterochromatin foci, and LAP2alpha protein levels were downregulated by proteasomal degradation. Although LAP2alpha was not required for the upregulation and reorganization of LINT-25 during cell cycle exit, transient expression of LINT-25 in proliferating cells caused loss of LAP2alpha and subsequent cell death. Our data show a role of LINT-25 and LAP2alpha during cell cycle exit, in which LINT-25 acts upstream of LAP2alpha.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/metabolismo , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Línea Celular , Células Cultivadas , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Células HeLa , Humanos , Immunoblotting , Inmunoprecipitación , Laminas/metabolismo , Proteínas de la Membrana/genética , Ratones , Microscopía Fluorescente , Proteínas Nucleares/genética , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Técnicas del Sistema de Dos Híbridos
11.
J Cell Biol ; 173(1): 83-93, 2006 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-16606692

RESUMEN

Lamina-associated polypeptide (LAP) 2alpha is a nonmembrane-bound LAP2 isoform that forms complexes with nucleoplasmic A-type lamins. In this study, we show that the overexpression of LAP2alpha in fibroblasts reduced proliferation and delayed entry into the cell cycle from a G0 arrest. In contrast, stable down-regulation of LAP2alpha by RNA interference accelerated proliferation and interfered with cell cycle exit upon serum starvation. The LAP2alpha-linked cell cycle phenotype is mediated by the retinoblastoma (Rb) protein because the LAP2alpha COOH terminus directly bound Rb, and overexpressed LAP2alpha inhibited E2F/Rb-dependent reporter gene activity in G1 phase in an Rb-dependent manner. Furthermore, LAP2alpha associated with promoter sequences in endogenous E2F/Rb-dependent target genes in vivo and negatively affected their expression. In addition, the expression of LAP2alpha in proliferating preadipocytes caused the accumulation of hypophosphorylated Rb, which is reminiscent of noncycling cells, and initiated partial differentiation into adipocytes. The effects of LAP2alpha on cell cycle progression and differentiation may be highly relevant for the cell- and tissue-specific phenotypes observed in laminopathic diseases.


Asunto(s)
Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción E2F/metabolismo , Proteínas de la Membrana/metabolismo , Lámina Nuclear/metabolismo , Proteína de Retinoblastoma/metabolismo , Células 3T3 , Adipocitos/metabolismo , Animales , Proliferación Celular , Medio de Cultivo Libre de Suero/farmacología , Proteínas de Unión al ADN/genética , Regulación hacia Abajo/fisiología , Factores de Transcripción E2F/genética , Fibroblastos/metabolismo , Regulación de la Expresión Génica/fisiología , Genes Reporteros/genética , Células HeLa , Humanos , Proteínas de la Membrana/genética , Ratones , Lámina Nuclear/ultraestructura , Regiones Promotoras Genéticas/genética , Estructura Terciaria de Proteína/fisiología , Interferencia de ARN , Elementos Reguladores de la Transcripción/genética , Fase de Descanso del Ciclo Celular/genética , Proteína de Retinoblastoma/genética
12.
Histochem Cell Biol ; 125(1-2): 33-41, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16142451

RESUMEN

The lamins are nuclear intermediate filament-type proteins forming the nuclear lamina meshwork at the inner nuclear membrane as well as complexes in the nucleoplasm. The recent discoveries that mutated A-type lamins and lamin-binding nuclear membrane proteins can be linked to numerous rare human diseases (laminopathies) affecting a multitude of tissues has changed the cell biologist's view of lamins as mere structural nuclear scaffold proteins. It is still unclear how mutations in these ubiquitously expressed proteins give rise to tissue-restricted pathological phenotypes. Potential disease models include mutation-caused defects in lamin structure and stability, the deregulation of gene expression, and impaired cell cycle control. This review brings together various previously proposed ideas and suggests a novel, more general, disease model based on an impairment of adult stem cell function and thus compromised tissue regeneration in laminopathic diseases.


Asunto(s)
Lamina Tipo A/fisiología , Regeneración/fisiología , Animales , Núcleo Celular/ultraestructura , Expresión Génica/fisiología , Humanos , Lamina Tipo A/genética , Regeneración/genética , Transducción de Señal/genética , Transducción de Señal/fisiología
13.
J Cell Sci ; 118(Pt 24): 5797-810, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16339967

RESUMEN

The LEM (lamina-associated polypeptide-emerin-MAN1) domain is a motif shared by a group of lamin-interacting proteins in the inner nuclear membrane (INM) and in the nucleoplasm. The LEM domain mediates binding to a DNA-crosslinking protein, barrier-to-autointegration factor (BAF). We describe a novel, ubiquitously expressed LEM domain protein, LEM2, which is structurally related to MAN1. LEM2 contains an N-terminal LEM motif, two predicted transmembrane domains and a MAN1-Src1p C-terminal (MSC) domain highly homologous to MAN1, but lacks the MAN1-specific C-terminal RNA-recognition motif. Immunofluorescence microscopy of digitonin-treated cells and subcellular fractionation identified LEM2 as a lamina-associated protein residing in the INM. LEM2 binds to the lamin C tail in vitro. Targeting of LEM2 to the nuclear envelope requires A-type lamins and is mediated by the N-terminal and transmembrane domains. Highly overexpressed LEM2 accumulates in patches at the nuclear envelope and forms membrane bridges between nuclei of adjacent cells. LEM2 structures recruit A-type lamins, emerin, MAN1 and BAF, whereas lamin B and lamin B receptor are excluded. Our data identify LEM2 as a novel A-type-lamin-associated INM protein involved in nuclear structure organization.


Asunto(s)
Lamina Tipo A/metabolismo , Lamina Tipo B/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Animales , Células COS , Chlorocebus aethiops , Proteínas de Unión al ADN , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lamina Tipo A/genética , Lamina Tipo B/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Membrana Nuclear/genética , Proteínas Nucleares/genética , Estructura Terciaria de Proteína/genética
14.
Hum Mutat ; 26(6): 566-74, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16247757

RESUMEN

Thymopoietin or TMPO (indicated by its alternative gene symbol, LAP2, in this work) has been proposed as a candidate disease gene for dilated cardiomyopathy (DCM), since a LAP2 product associates with nucleoplasmic lamins A/C, which are encoded by the DCM gene LMNA. We developed a study to screen for genetic mutations in LAP2 in a large collection of DCM patients and families. A total of 113 subjects from 88 families (56 with familial DCM (FDC) and 32 with sporadic DCM) were screened for LAP2 mutations using denaturing high-performance liquid chromatography and sequence analysis. We found a single putative mutation affecting the LAP2alpha isoform in one FDC pedigree. The mutation predicts an Arg690Cys substitution (c.2068C>T; p.R690C) located in the C-terminal domain of the LAP2alpha protein, a region that is known to interact with lamin A/C. RT-PCR, Western blot analyses, and immunolocalization revealed low-level LAP2alpha expression in adult cardiac muscle, and localization to a subset of nuclei. Mutated Arg690Cys LAP2alpha expressed in HeLa cells localized to the nucleoplasm like wild-type LAP2alpha, with no effect on peripheral and nucleoplasmic lamin A distribution. However, the in vitro interaction of mutated LAP2alpha with the pre-lamin A C-terminus was significantly compromised compared to the wild-type protein. LAP2 mutations may represent a rare cause of DCM. The Arg690Cys mutation altered the observed LAP2alpha interaction with A-type lamins. Our finding implicates a novel nuclear lamina-associated protein in the pathogenesis of genetic forms of dilated cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/genética , Proteínas de Unión al ADN/genética , Predisposición Genética a la Enfermedad , Proteínas de la Membrana/genética , Mutación Missense , Cromatografía Liquida , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Femenino , Pruebas Genéticas , Células HeLa , Humanos , Lamina Tipo A/química , Lamina Tipo A/metabolismo , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Miocardio/citología , Miocardio/metabolismo , Linaje , Isoformas de Proteínas/genética , Estructura Terciaria de Proteína
15.
J Cell Sci ; 118(Pt 15): 3419-30, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16079285

RESUMEN

Nesprins form a novel class of nuclear envelope-anchored spectrin-repeat proteins. We show that a direct association of their highly conserved C-terminal luminal domain with the inner nuclear membrane protein Sun1 mediates their nuclear envelope localisation. In Nesprin-1 and Nesprin-2 the conserved C-terminal amino acids PPPX are essential for the interaction with a C-terminal region in Sun1. In fact, Sun1 is required for the proper nuclear envelope localisation of Nesprin-2 as shown using dominant-negative mutants and by knockdown of Sun1 expression. Sun1 itself does not require functional A-type lamins for its localisation at the inner nuclear membrane in mammalian cells. Our findings propose a conserved nuclear anchorage mechanism between Caenorhabditis elegans and mammals and suggest a model in which Sun1 serves as a ;structural bridge' connecting the nuclear interior with the actin cytoskeleton.


Asunto(s)
Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Animales , Células COS , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Núcleo Celular/metabolismo , Chlorocebus aethiops , Clonación Molecular , Proteínas del Citoesqueleto , Células HeLa , Humanos , Membranas Intracelulares/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas Asociadas a Microtúbulos/genética , Modelos Biológicos , Proteínas del Tejido Nervioso/genética , Membrana Nuclear/clasificación , Membrana Nuclear/ultraestructura , Proteínas Nucleares/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
16.
Carcinogenesis ; 26(5): 931-42, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15705598

RESUMEN

Immortalized p19(ARF) null hepatocytes (MIM) feature a high degree of functional differentiation and are susceptible to transforming growth factor (TGF)-beta driven growth arrest and apoptosis. In contrast, polarized MIM hepatocytes expressing hyperactive Ha-Ras continue proliferation in cooperation with TGF-beta, and adopt an invasive phenotype by executing an epithelial to mesenchymal transition (EMT). In this study, we analyzed the involvement of Ras subeffectors in TGF-beta mediated hepatocellular EMT by employing MIM hepatocytes, which express Ras mutants allowing selective activation of either mitogen-activated protein kinase (MAPK) signaling (V12-S35) or phosphoinositide 3-OH (PI3)3 kinase (PI3K) signaling (V12-C40). We found that MAPK signaling in MIM-S35 hepatocytes was necessary and sufficient to promote resistance to TGF-beta mediated inhibition of proliferation in vitro and in vivo. MIM-S35 hepatocytes showed also PI3K activation during EMT, however, MAPK signaling on its own protected hepatocytes from apoptosis. Yet, MIM-C40 hepatocytes failed to form tumors and required additional MAPK stimulation to overcome TGF-beta mediated growth arrest. In vivo, the collaboration of MAPK signaling and TGF-beta activity drastically accelerated the cell-cycle progression of the hepatocytes, leading to vast tumor formation. From these data we conclude that MAPK is crucial for the cooperation with TGF-beta to regulate the proliferation as well as the survival of hepatocytes during EMT, and causes the fatal increase in hepatocellular tumor progression.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Proteínas ras/metabolismo , Animales , Transformación Celular Neoplásica/metabolismo , Humanos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Invasividad Neoplásica
17.
Oncogene ; 23(15): 2672-2680, 2004 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-14755243

RESUMEN

Several signalling pathways contribute to the regulation of epithelial to mesenchymal transition (EMT), either during developmentally regulated processes or in cancer progression and metastasis. Induction of EMT in fully polarized mouse mammary epithelial cells (EpH4) by an inducible c-fos estrogen receptor (FosER) oncoprotein involves loss of E-cadherin expression, nuclear translocation of beta-catenin, and autocrine production of TGFbeta. Reporter assays demonstrate that both beta-catenin/LEF-TCF- and TGFbeta-Smad-dependent signalling activities are upregulated, probably coregulating mesenchymal-specific gene expression during EMT. Stable expression of E-cadherin in mesenchymal FosER cells decreased beta-catenin activity and reduced cell proliferation. However, these cells still exhibited a defect in epithelial polarization and expressed E-cadherin/beta-catenin complexes in the entire plasma membrane. On the other hand, inhibition of TGFbeta-Smad signalling in mesenchymal FosER cells induced flat, cobblestone-like clusters of cells, which relocalized beta-catenin to the plasma membrane but still lacked detectable E-cadherin. Interestingly, inhibition of TGFbeta signalling in the E-cadherin-expressing mesenchymal FosER cells caused their reversion to a polarized epithelial phenotype, in which E-cadherin, beta-catenin, and ZO-1 were localized at their correct lateral plasma membrane domains. These results demonstrate that loss of E-cadherin can contribute to increased LEF/TCF-beta-catenin signalling, which in turn cooperates with autocrine TGFbeta signalling to maintain an undifferentiated mesenchymal phenotype.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Epitelio/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de Estrógenos/metabolismo , Transducción de Señal , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Transporte Activo de Núcleo Celular , Animales , Cadherinas/metabolismo , Adhesión Celular , Diferenciación Celular , División Celular , Línea Celular , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , ADN Complementario/metabolismo , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Genes Reporteros , Mesodermo/patología , Ratones , Microscopía Fluorescente , Neoplasias/metabolismo , Fenotipo , Isoformas de Proteínas , Estructura Terciaria de Proteína , ARN/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Activación Transcripcional , Transfección , Regulación hacia Arriba , beta Catenina
18.
Mutat Res ; 566(1): 9-20, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14706509

RESUMEN

Carcinomas arising from epithelial cells represent the most prevalent malignancies in humans, and metastasis is the major cause for the death of carcinoma patients. The breakdown of epithelial cell homeostasis leading to aggressive cancer progression has been correlated with the loss of epithelial characteristics and the acquisition of a migratory phenotype. This phenomenon, referred to as epithelial to mesenchymal transition (EMT), is considered as a crucial event in late stage tumorigenesis. Here we summarize the multitude of EMT models derived from different tissues, and review the diversity of molecular mechanisms contributing to the plasticity of epithelial cells. In particular, the synergism between activation of Ras, provided by the aberrant stimulation of receptor tyrosine kinases, and transforming growth factor (TGF)-beta signaling plays a pivotal role in inducing EMT of various epithelial cell types. Cytokines such as TGF-beta and extracellular matrix molecules are thought to fundamentally contribute to the microenvironmental interaction between stromal and malignant cells, and provide the basis for a broad repertoire of epithelial differentiation. Investigations of EMT tumor models, which represent in vitro correlates to local invasion and metastasis in vivo, facilitate the identification of diagnostic markers for a more accurate and faithful clinical and pathological assessment of epithelial tumors. In addition, the analysis of molecular mechanisms involved in EMT might yield novel therapeutic targets for the specific treatment of aggressive carcinomas.


Asunto(s)
Transformación Celular Neoplásica , Células Epiteliales/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias Glandulares y Epiteliales/patología , Animales , Humanos , Modelos Biológicos , Transducción de Señal
19.
Oncogene ; 22(43): 6725-38, 2003 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-14555986

RESUMEN

Hepatocytes adopt an invasive and metastatic phenotype caused by the cooperation of transforming growth factor (TGF)-beta and oncogenic Ha-Ras. In the initial phase of this process, c-Fos is rapidly induced by TGF-beta, but then decreases to undetectable levels. Here, we investigated the functional implications of c-Fos activation and its contribution to hepatocellular tumorigenesis. By employing conditional c-Fos expression, we observed that continuous activation of c-Fos and consequently AP-1 activity leads to depolarization of differentiated murine epithelial hepatocytes. Most remarkably, this change in morphology was associated with inhibition of proliferation and induction of cell death. Coexpression of antiapoptotic Bcl-XL or scavenging of reactive oxygen species was sufficient to prevent the c-Fos-mediated phenotype. In contrast, the cooperation of c-Fos with oncogenic Ha-Ras or a Ras mutant selectively activating the MAPK pathway even enhanced c-Fos-induced effects. Showing the negative role in hepatocellular tumorigenesis, c-Fos repressed oncogenic Ras-driven anchorage-independent growth in vitro and strongly suppressed tumour formation in vivo. Taken together, we demonstrate that c-Fos modulates plasticity of epithelial hepatocytes and acts tumour suppressive in neoplastic hepatocytes by stimulating cell cycle inhibition and cell death.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Apoptosis , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Muerte Celular , División Celular , Línea Celular , Supervivencia Celular , Transformación Celular Neoplásica , Células Epiteliales , Citometría de Flujo , Genes ras , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos BALB C , Fenotipo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno , Transducción de Señal , Factores de Tiempo , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción CHOP , Factores de Transcripción/metabolismo , Transfección , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba , Proteína bcl-X
20.
Exp Cell Res ; 283(2): 184-95, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12581738

RESUMEN

TLS/FUS is a nucleic acid-binding protein whose N-terminal half functions as a transcriptional activator domain in fusion oncoproteins found in human leukemias and liposarcomas. Previous reports have suggested a role for TLS/FUS in transcription and splicing processes. Here we report the association of TLS/FUS with the nuclear matrix and investigate its role in splicing. Splicing of two pre-mRNAs was inhibited in a TLS/FUS-immunodepleted extract and could only be partly restored by addition of recombinant TLS/FUS or/and SR proteins, known interaction partners of TLS/FUS. The subsequent analysis of TLS/FUS immunoprecipitates revealed that, in addition to the SR proteins SC35 and SRp75, the splicing factor PTB (hnRNPI) and the splicing coactivator SRm160 are complexed with TLS/FUS, thus explaining the inability to restore splicing completely. Coimmunolocalization confirmed the nuclear matrix association and interaction of TLS/FUS with PTB, SR proteins, and SRm160. Our results suggest that the matrix protein TLS/FUS plays a role in spliceosome assembly.


Asunto(s)
Matriz Nuclear/química , Empalme del ARN , Proteína FUS de Unión a ARN/fisiología , Antígenos Nucleares/metabolismo , Células HeLa , Humanos , Microscopía Fluorescente , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Pruebas de Precipitina , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Proteína FUS de Unión a ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Partícula de Reconocimiento de Señal/metabolismo , Empalmosomas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...