Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Neurobiol ; 58(6): 2523-2541, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33459966

RESUMEN

Sigma-1 receptors (Sig-1Rs) are endoplasmic reticulum (ER) chaperones implicated in neuropathic pain. Here we examine if the Sig-1R may relate to neuropathic pain at the level of dorsal root ganglia (DRG). We focus on the neuronal excitability of DRG in a "spare nerve injury" (SNI) model of neuropathic pain in rats and find that Sig-1Rs likely contribute to the genesis of DRG neuronal excitability by decreasing the protein level of voltage-gated Cav2.2 as a translational inhibitor of mRNA. Specifically, during SNI, Sig-1Rs translocate from ER to the nuclear envelope via a trafficking protein Sec61ß. At the nucleus, the Sig-1R interacts with cFos and binds to the promoter of 4E-BP1, leading to an upregulation of 4E-BP1 that binds and prevents eIF4E from initiating the mRNA translation for Cav2.2. Interestingly, in Sig-1R knockout HEK cells, Cav2.2 is upregulated. In accordance with those findings, we find that intra-DRG injection of Sig-1R agonist (+)pentazocine increases frequency of action potentials via regulation of voltage-gated Ca2+ channels. Conversely, intra-DRG injection of Sig-1R antagonist BD1047 attenuates neuropathic pain. Hence, we discover that the Sig-1R chaperone causes neuropathic pain indirectly as a translational inhibitor.


Asunto(s)
Genoma , Neuralgia/genética , Receptores sigma/metabolismo , Animales , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Retículo Endoplásmico/metabolismo , Factor 4E Eucariótico de Iniciación/metabolismo , Ganglios Espinales/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Tejido Nervioso/lesiones , Tejido Nervioso/patología , Membrana Nuclear/metabolismo , Regiones Promotoras Genéticas/genética , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Caperuzas de ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptores sigma/agonistas , Receptores sigma/genética , Canales de Translocación SEC/metabolismo , Transcripción Genética , Receptor Sigma-1
2.
Cells ; 7(5)2018 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-29772801

RESUMEN

The LMNA gene encodes lamins A and C with key roles in nuclear structure, signaling, gene regulation, and genome integrity. Mutations in LMNA cause over 12 diseases ('laminopathies'). Lamins A and C are identical for their first 566 residues. However, they form separate filaments in vivo, with apparently distinct roles. We report that lamin A is ß-O-linked N-acetylglucosamine-(O-GlcNAc)-modified in human hepatoma (Huh7) cells and in mouse liver. In vitro assays with purified O-GlcNAc transferase (OGT) enzyme showed robust O-GlcNAcylation of recombinant mature lamin A tails (residues 385⁻646), with no detectable modification of lamin B1, lamin C, or 'progerin' (Δ50) tails. Using mass spectrometry, we identified 11 O-GlcNAc sites in a 'sweet spot' unique to lamin A, with up to seven sugars per peptide. Most sites were unpredicted by current algorithms. Double-mutant (S612A/T643A) lamin A tails were still robustly O-GlcNAc-modified at seven sites. By contrast, O-GlcNAcylation was undetectable on tails bearing deletion Δ50, which causes Hutchinson⁻Gilford progeria syndrome, and greatly reduced by deletion Δ35. We conclude that residues deleted in progeria are required for substrate recognition and/or modification by OGT in vitro. Interestingly, deletion Δ35, which does not remove the majority of identified O-GlcNAc sites, does remove potential OGT-association motifs (lamin A residues 622⁻625 and 639⁻645) homologous to that in mouse Tet1. These biochemical results are significant because they identify a novel molecular pathway that may profoundly influence lamin A function. The hypothesis that lamin A is selectively regulated by OGT warrants future testing in vivo, along with two predictions: genetic variants may contribute to disease by perturbing OGT-dependent regulation, and nutrient or other stresses might cause OGT to misregulate wildtype lamin A.

3.
Nucleus ; 4(4): 298-314, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23873439

RESUMEN

Emerin, a conserved LEM-domain protein, is among the few nuclear membrane proteins for which extensive basic knowledge--biochemistry, partners, functions, localizations, posttranslational regulation, roles in development and links to human disease--is available. This review summarizes emerin and its emerging roles in nuclear "lamina" structure, chromatin tethering, gene regulation, mitosis, nuclear assembly, development, signaling and mechano-transduction. We also highlight many open questions, exploration of which will be critical to understand how this intriguing nuclear membrane protein and its "family" influence the genome.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de la Membrana/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Animales , Humanos , Proteínas de la Membrana/genética , Proteínas Nucleares/genética
4.
PLoS One ; 7(9): e45819, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029259

RESUMEN

GAGE proteins are highly similar, primate-specific molecules with unique primary structure and undefined cellular roles. They are restricted to cells of the germ line in adult healthy individuals, but are broadly expressed in a wide range of cancers. In a yeast two-hybrid screen we identified the metazoan transcriptional regulator, Germ cell-less (GCL), as an interaction partner of GAGE12I. GCL directly binds LEM-domain proteins (LAP2ß, emerin, MAN1) at the nuclear envelope, and we found that GAGE proteins were recruited to the nuclear envelope inner membrane by GCL. Based on yeast two-hybrid analysis and pull-down experiments of GCL polypeptides, GCL residues 209-320 (which includes the BACK domain) were deduced sufficient for association with GAGE proteins. GAGE mRNAs and GCL mRNA were demonstrated in human testis and most types of cancers, and at the protein level GAGE members and GCL were co-expressed in cancer cell lines. Structural studies of GAGE proteins revealed no distinct secondary or tertiary structure, suggesting they are intrinsically disordered. Interestingly GAGE proteins formed stable complexes with dsDNA in vitro at physiological concentrations, and GAGE12I bound several different dsDNA fragments, suggesting sequence-nonspecific binding. Dual association of GAGE family members with GCL at the nuclear envelope inner membrane in cells, and with dsDNA in vitro, implicate GAGE proteins in chromatin regulation in germ cells and cancer cells.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Proteínas de Neoplasias/metabolismo , Membrana Nuclear/metabolismo , Factores de Transcripción/metabolismo , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Línea Celular , Transformación Celular Neoplásica/metabolismo , Cromatina/metabolismo , Dicroismo Circular , ADN/química , Ensayo de Cambio de Movilidad Electroforética , Expresión Génica , Humanos , Masculino , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Especificidad de Órganos , Plásmidos/química , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Testículo/metabolismo , Factores de Transcripción/genética , Técnicas del Sistema de Dos Híbridos
5.
Cancer Epidemiol Biomarkers Prev ; 21(12): 2159-66, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23064005

RESUMEN

BACKGROUND: Leukemia is a common cancer among U.S. adults but there are few established risk factors. If leukemia risks are substantially influenced by exposures that vary in prevalence across generations, then population incidence rates should vary significantly by birth cohort. However, prior studies have not examined leukemia birth cohort effects using contemporary data and methods. METHODS: We used incidence data from the National Cancer Institute's Surveillance, Epidemiology and End Results Program from 1992 through 2009 for adults 25-84 years old and age period cohort models to estimate incidence rate ratios according to birth cohort for acute myeloid leukemia (AML), acute lymphoid leukemia (ALL), chronic myeloid leukemia (CML), and chronic lymphoid leukemia (CLL). RESULTS: Leukemia incidence varied significantly between birth cohorts for each major leukemia type in men and women except female AMLs; changes on the order of 1% per birth year or 20% per generation were observed. The most significant birth cohort signatures were observed for CLLs and AMLs in men, which were decreasing and increasing, respectively, in cohorts born since 1946. CONCLUSIONS: Our results support the hypothesis that adult leukemia risks are significantly modulated by environmental and lifestyle exposures. IMPACT: A number of well-established (smoking, certain chemicals, radiation) and newly recognized (obesity) leukemia risk factors are modifiable; ultimately, efforts to promote healthy lifestyles might also help reduce incidence rates of adult leukemia.


Asunto(s)
Leucemia/epidemiología , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Factores de Riesgo , Programa de VERF , Estados Unidos/epidemiología
6.
J Cell Sci ; 122(Pt 20): 3780-90, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19789182

RESUMEN

X-linked recessive Emery-Dreifuss muscular dystrophy (EDMD) is caused by loss of emerin, a nuclear-membrane protein with roles in nuclear architecture, gene regulation and signaling. Phosphoproteomic studies have identified 13 sites of tyrosine phosphorylation in emerin. We validated one study, confirming that emerin is hyper-tyrosine-phosphorylated in Her2-overexpressing cells. We discovered that non-receptor tyrosine kinases Src and Abl each phosphorylate emerin and a related protein, LAP2beta, directly. Src phosphorylated emerin specifically at Y59, Y74 and Y95; the corresponding triple Y-to-F (;FFF') mutation reduced tyrosine phosphorylation by approximately 70% in vitro and in vivo. Substitutions that removed a single hydroxyl moiety either decreased (Y19F, Y34, Y161F) or increased (Y4F) emerin binding to BAF in cells. Y19F, Y34F, Y161F and the FFF mutant also reduced recombinant emerin binding to BAF from HeLa lysates, demonstrating the involvement of both LEM-domain and distal phosphorylatable tyrosines in binding BAF. We conclude that emerin function is regulated by multiple tyrosine kinases, including Her2, Src and Abl, two of which (Her2, Src) regulate striated muscle. These findings suggest roles for emerin as a downstream effector and ;signal integrator' for tyrosine kinase signaling pathway(s) at the nuclear envelope.


Asunto(s)
Proteínas de la Membrana/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Fosfotirosina/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Familia-src Quinasas/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Animales , Células HeLa , Humanos , Espectrometría de Masas , Proteínas de la Membrana/química , Ratones , Datos de Secuencia Molecular , Mutación Missense/genética , Células 3T3 NIH , Proteínas Nucleares/química , Receptor ErbB-2/metabolismo , Transducción de Señal
7.
PLoS One ; 4(9): e7050, 2009 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-19759913

RESUMEN

Nuclear lamin filaments and associated proteins form a nucleoskeletal ("lamina") network required for transcription, replication, chromatin organization and epigenetic regulation in metazoans. Lamina defects cause human disease ("laminopathies") and are linked to aging. Barrier-to-autointegration factor (BAF) is a mobile and essential component of the nuclear lamina that binds directly to histones, lamins and LEM-domain proteins, including the inner nuclear membrane protein emerin, and has roles in chromatin structure, mitosis and gene regulation. To understand BAF's mechanisms of action, BAF associated proteins were affinity-purified from HeLa cell nuclear lysates using BAF-conjugated beads, and identified by tandem mass spectrometry or independently identified and quantified using the iTRAQ method. We recovered A- and B-type lamins and core histones, all known to bind BAF directly, plus four human transcription factors (Requiem, NonO, p15, LEDGF), disease-linked proteins (e.g., Huntingtin, Treacle) and several proteins and enzymes that regulate chromatin. Association with endogenous BAF was independently validated by co-immunoprecipitation from HeLa cells for seven candidates including Requiem, poly(ADP-ribose) polymerase 1 (PARP1), retinoblastoma binding protein 4 (RBBP4), damage-specific DNA binding protein 1 (DDB1) and DDB2. Interestingly, endogenous BAF and emerin each associated with DDB2 and CUL4A in a UV- and time-dependent manner, suggesting BAF and emerin have dynamic roles in genome integrity and might help couple DNA damage responses to the nuclear lamina network. We conclude this proteome is a rich source of candidate partners for BAF and potentially also A- and B-type lamins, which may reveal how chromatin regulation and genome integrity are linked to nuclear structure.


Asunto(s)
Cromatina/química , Proteínas de la Membrana/metabolismo , Lámina Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Proteoma/química , Secuencia de Aminoácidos , Proteínas Cullin/metabolismo , Proteínas de Unión al ADN/metabolismo , Células HeLa , Histonas/química , Humanos , Datos de Secuencia Molecular , Poli(ADP-Ribosa) Polimerasas/química , Estructura Terciaria de Proteína , Proteína 4 de Unión a Retinoblastoma/metabolismo , Homología de Secuencia de Aminoácido
8.
Biochemistry ; 46(30): 8897-908, 2007 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-17620012

RESUMEN

Using recombinant bead-conjugated emerin, we affinity-purified seven proteins from HeLa cell nuclear lysates that bind emerin either directly or indirectly. These proteins were identified by mass spectrometry as nuclear alphaII-spectrin, nonmuscle myosin heavy chain alpha, Lmo7 (a predicted transcription regulator; reported separately), nuclear myosin I, beta-actin (reported separately), calponin 3, and SIKE. We now report that emerin binds nuclear myosin I (NMI, a molecular motor) directly in vitro. Furthermore, bead-conjugated emerin bound nuclear alphaII-spectrin and NMI equally well with or without ATP (which stimulates motor activity), whereas ATP decreased actin binding by 65%. Thus alphaII-spectrin and NMI interact stably with emerin. To investigate the physiological relevance of these interactions, we used antibodies against emerin to affinity-purify emerin-associated protein complexes from HeLa cells and then further purified by ion-exchange chromatography to resolve by net charge and by size exclusion chromatography yielding six distinct emerin-containing fractions (0.5-1.6 MDa). Western blotting suggested that each complex had distinct components involved in nuclear architecture (e.g., NMI, alphaII-spectrin, lamins) or gene or chromatin regulation (BAF, transcription regulators, HDACs). Additional constituents were identified by mass spectrometry. One putative gene-regulatory complex (complex 32) included core components of the nuclear corepressor (NCoR) complex, which mediates gene regulation by thyroid hormone and other nuclear receptors. When expressed in HeLa cells, FLAG-tagged NCoR subunits Gps2, HDAC3, TBLR1, and NCoR each co-immunoprecipitated emerin, validating one putative complex. These findings support the hypothesis that emerin scaffolds a variety of functionally distinct multiprotein complexes at the nuclear envelope in vivo. Notably included are nuclear myosin I-containing complexes that might sense and regulate mechanical tension at the nuclear envelope.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/fisiología , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/fisiología , Proteoma/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Sitios de Unión/fisiología , Línea Celular Tumoral , Cromatina/metabolismo , Cromatografía de Afinidad , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Células HeLa , Humanos , Inmunoprecipitación , Proteínas de la Membrana/química , Proteínas de la Membrana/aislamiento & purificación , Proteínas Asociadas a Matriz Nuclear , Proteínas Nucleares/química , Proteínas Nucleares/aislamiento & purificación , Unión Proteica/fisiología , Biosíntesis de Proteínas , Proteoma/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo
9.
Exp Cell Res ; 312(4): 478-87, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16337940

RESUMEN

Barrier-to-autointegration factor (BAF) is an essential chromatin protein conserved in metazoans. BAF has roles in nuclear assembly, chromatin organization, gene expression, and gonad development and is exploited by retroviruses. BAF forms stable dimers that bind nonspecifically to dsDNA and specifically to LEM-domain proteins (e.g., LAP2beta, emerin, MAN1), homeodomain transcription factors, histones, and lamin A. We characterized a protein named BAF-Like (BAF-L) that in humans is 40% identical to BAF. Overexpression studies in HeLa cells show that BAF-L, like BAF, is a predominantly nuclear protein. Recombinant BAF-L forms stable homodimers and heterodimerizes with BAF in vitro and also interacts with BAF in vivo. BAF-L does not bind significantly to DNA, LAP2beta, or emerin but can form ternary complexes in vitro with BAF plus DNA, or BAF plus LAP2beta. Levels of BAF-L mRNA were high in pancreas and testis, suggesting functions in the germline. BAF-L mRNA was detectable at low levels in eleven other tissues and undetectable in heart and skeletal muscle which are specifically affected by Emery-Dreifuss muscular dystrophy, a disease caused by mutations in either emerin or lamin A. We propose that BAF-L regulates BAF function via heterodimerization and might thereby influence tissue-specific roles of BAF.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Secuencia de Aminoácidos , Animales , Núcleo Celular/metabolismo , ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Dimerización , Técnica del Anticuerpo Fluorescente Indirecta , Perfilación de la Expresión Génica , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular , Masculino , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Páncreas/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido , Testículo/metabolismo , Timopoyetinas/metabolismo , Transfección
10.
J Biol Chem ; 280(14): 13863-70, 2005 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-15681850

RESUMEN

MAN1 is a vertebrate nuclear inner membrane protein that inhibits Smad signaling downstream of transforming growth factor beta. MAN1 has an exposed LEM domain-containing N-terminal region ("MAN1-N"), two transmembrane domains, and an exposed C-terminal domain ("MAN1-C"). Many regions of human MAN1 are homologous to emerin, a LEM domain nuclear protein, loss of which causes Emery-Dreifuss muscular dystrophy (EDMD). To test the hypothesis that MAN1 function might overlap with emerin, we tested different polypeptide fragments of MAN1 for binding to selected partners of emerin. Our findings support this hypothesis. Blot overlay assays and co-immunoprecipitation studies showed that MAN1-C binds the transcription regulators GCL, Btf, and barrier-to-autointegration factor (BAF). BAF binding to this region, which has no LEM domain, was notable. Sequence alignments identified a potential BAF-binding motif, characterized by the conserved residues Ser-Arg-Val, in MAN1-C and two other BAF-binding proteins. The other region, MAN1-N, bound directly to BAF, lamin A, and lamin B1, supporting functional overlap with emerin. Unexpectedly, three independent assays showed that MAN1-N also bound directly to emerin. Proposed MAN1-emerin complexes are discussed in the context of EDMD disease mechanisms and potential in vivo functions.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/metabolismo , Timopoyetinas/metabolismo , Secuencia de Aminoácidos , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Timopoyetinas/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
11.
Trends Cell Biol ; 14(5): 261-6, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15130582

RESUMEN

Barrier-to-autointegration factor (BAF) is an essential protein that is highly conserved in metazoan evolution. BAF binds directly to double-stranded DNA, nuclear LEM-domain proteins, lamin A and transcription activators. BAF is also a host cell component of retroviral pre-integration complexes. BAF binds matrix, a retroviral protein, and facilitates efficient retroviral DNA integration in vitro through unknown mechanisms. New findings suggest that BAF has structural roles in nuclear assembly and chromatin organization, represses gene expression and might interlink chromatin structure, nuclear architecture and gene regulation in metazoans.


Asunto(s)
Núcleo Celular/química , Núcleo Celular/metabolismo , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Retroviridae/fisiología , Integración Viral/fisiología , Secuencia de Aminoácidos , Animales , Proteínas de Unión al ADN/química , Humanos , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Represoras/metabolismo
12.
J Struct Biol ; 147(1): 31-41, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15109603

RESUMEN

Barrier-to-autointegration factor (BAF) is a conserved 10 kDa DNA-binding protein. BAF interacts with LEM-domain proteins including emerin, LAP2 beta, and MAN1 in the inner nuclear membrane. Using fluorescence recovery after photobleaching (FRAP) and fluorescence loss in photobleaching (FLIP), we compared the mobility of BAF to its partners emerin, LAP2 beta, and MAN1 in living HeLa cells. Like endogenous BAF, GFP-BAF was enriched at the nuclear envelope, and found inside the nucleus and in the cytoplasm during interphase. At every location, FRAP and FLIP analysis showed that GFP-BAF diffused rapidly; the halftimes for recovery in a 0.8 microm square area were 260 ms at the nuclear envelope, and even faster inside the nucleus and in the cytoplasm. GFP-fused emerin, LAP2 beta, and MAN1 were all relatively immobile, with recovery halftimes of about 1 min, for a 2 microm square area. Thus, BAF is dynamic and mobile during interphase, in stark contrast to its nuclear envelope partners. FLIP results further showed that rapidly diffusing cytoplasmic and nuclear pools of GFP-BAF were distinctly regulated, with nuclear GFP-BAF unable to replenish cytoplasmic BAF. Fluorescence resonance energy transfer (FRET) results showed that CFP-BAF binds directly to YFP-emerin at the inner nuclear membrane of living cells. We propose a "touch-and-go" model in which BAF binds emerin frequently but transiently during interphase. These findings contrast with the slow mobility of both GFP-BAF and GFP-emerin during telophase, when they colocalized at the 'core' region of telophase chromosomes at early stages of nuclear assembly.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo , Transferencia Resonante de Energía de Fluorescencia , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/metabolismo , Timopoyetinas/metabolismo , Animales , Proteínas de Unión al ADN/genética , Recuperación de Fluorescencia tras Fotoblanqueo/métodos , Transferencia Resonante de Energía de Fluorescencia/métodos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Proteínas de la Membrana/genética , Membrana Nuclear/metabolismo , Proteínas Nucleares/genética , Unión Proteica , Transporte de Proteínas/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Timopoyetinas/genética
13.
Eur J Biochem ; 271(5): 1035-45, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15009215

RESUMEN

Loss of functional emerin, a nuclear membrane protein, causes X-linked recessive Emery-Dreifuss muscular dystrophy. In a yeast two-hybrid screen, we found that emerin interacts with Btf, a death-promoting transcriptional repressor, which is expressed at high levels in skeletal muscle. Biochemical analysis showed that emerin binds Btf with an equilibrium affinity (KD) of 100 nm. Using a collection of 21 clustered alanine-substitution mutations in emerin, the residues required for binding to Btf mapped to two regions of emerin that flank its lamin-binding domain. Two disease-causing mutations in emerin, S54F and Delta95-99, disrupted binding to Btf. The Delta95-99 mutation was relatively uninformative, as this mutation also disrupts emerin binding to lamin A and a different transcription repressor named germ cell-less (GCL). In striking contrast, emerin mutant S54F, which binds normally to barrier-to-autointegration factor, lamin A and GCL, selectively disrupted emerin binding to Btf. We localized endogenous Btf in HeLa cells by indirect immunoflurorescence using affinity-purified antibodies against Btf. In nonapoptotic HeLa cells Btf was found in dot-like structures throughout the nuclear interior. However, within 3 h after treating cells with Fas antibody to induce apoptosis, the distribution of Btf changed, and Btf concentrated in a distinct zone near the nuclear envelope. These results suggest that Btf localization is regulated by apoptotic signals, and that loss of emerin binding to Btf may be relevant to muscle wasting in Emery-Dreifuss muscular dystrophy.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/metabolismo , Mutación Missense , Proteínas Represoras/metabolismo , Timopoyetinas/genética , Timopoyetinas/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Apoptosis/fisiología , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Membrana Nuclear/metabolismo , Proteínas Nucleares , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Represoras/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Técnicas del Sistema de Dos Híbridos
14.
J Cell Sci ; 117(Pt 7): 979-87, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14996929

RESUMEN

What do such diverse molecules as DNA, actin, retinoblastoma protein and protein kinase Calpha all have in common? They and additional partners bind 'A-type' lamins, which form stable filaments in animal cell nuclei. Mutations in A-type lamins cause a bewildering range of tissue-specific diseases, termed 'laminopathies', including Emery-Dreifuss muscular dystrophy and the devastating Hutchinson-Gilford progeria syndrome, which mimics premature aging. Considered individually and collectively, partners for A-type lamins form four loose groups: architectural partners, chromatin partners, gene-regulatory partners and signaling partners. We describe 16 partners in detail, summarize their binding sites in A-type lamins, and sketch portraits of ternary complexes and functional pathways that might depend on lamins in vivo. On the basis of our limited current knowledge, we propose lamin-associated complexes with multiple components relevant to nuclear structure (e.g. emerin, nesprin 1alpha, actin) or signaling and gene regulation (e.g. LAP2alpha, retinoblastoma, E2F-DP heterodimers, genes) as 'food for thought'. Testing these ideas will deepen our understanding of nuclear function and human disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Lamina Tipo A/metabolismo , Animales , Sitios de Unión , Proteínas Portadoras/química , Núcleo Celular/metabolismo , Cromatina/metabolismo , Regulación de la Expresión Génica , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Humanos , Lamina Tipo A/química , Lamina Tipo A/genética , Sustancias Macromoleculares , Modelos Biológicos , Mutación , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Unión Proteica , Transducción de Señal
15.
J Virol ; 77(24): 13084-92, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14645565

RESUMEN

Barrier-to-autointegration factor (BAF) is a conserved human chromatin protein exploited by retroviruses. Previous investigators showed that BAF binds double-stranded DNA nonspecifically and is a host component of preintegration complexes (PICs) isolated from cells infected with human immunodeficiency virus type 1 (HIV-1) or Moloney murine leukemia virus. BAF protects PIC structure and stimulates the integration of salt-stripped PICs into target DNA in vitro. PICs are thought to acquire BAF from the cytoplasm during infection. However, we identified two human tissues (of 16 tested) in which BAF mRNA was not detected: thymus and peripheral blood leukocytes, which are enriched in CD4(+) T lymphocytes and macrophage precursors, respectively. BAF protein was detected in activated but not resting CD4(+) T lymphocytes; thus, if BAF were essential for PIC function, we hypothesized that virions might "bring their own BAF." Supporting this model, BAF copurified with HIV-1 virions that were digested with subtilisin to remove microvesicle contaminants, and BAF was present in approximately zero to three copies per virion. In three independent assays, BAF bound directly to both p55 Gag (the structural precursor of HIV-1 virions) and its cleaved product, matrix. Using lysates from cells overexpressing Gag, endogenous BAF and Gag were coimmunoprecipitated by antibodies against Gag. Purified recombinant BAF had low micromolar affinities (1.1 to 1.4 micro M) for recombinant Gag and matrix. We conclude that BAF is present at low levels in incoming virions, in addition to being acquired from the cytoplasm of newly infected cells. We further conclude that BAF might contribute to the assembly or activity of HIV-1 PICs through direct binding to matrix, as well as DNA.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Productos del Gen gag/metabolismo , Antígenos VIH/metabolismo , VIH-1/metabolismo , Proteínas Nucleares/metabolismo , Precursores de Proteínas/metabolismo , Proteínas Virales , Virión/metabolismo , Integración Viral , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proteínas de Unión al ADN/genética , Productos del Gen gag/genética , VIH-1/patogenicidad , Células HeLa , Humanos , Proteínas Nucleares/genética , Precursores de Proteínas/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana
16.
Eur J Biochem ; 270(11): 2459-66, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12755701

RESUMEN

Emerin is a nuclear membrane protein that interacts with lamin A/C at the nuclear envelope. Mutations in either emerin or lamin A/C cause Emery-Dreifuss muscular dystrophy (EDMD). The functions of emerin are poorly understood, but EDMD affects mainly skeletal and cardiac muscle. We used a high-stringency yeast two-hybrid method to screen a human heart cDNA library, with full-length emerin as bait. Four out of five candidate interactors identified were nuclear proteins: lamin A, splicing factor YT521-B, proteasome subunit PA28 gamma and transcription factor vav-1. Specific binding between emerin and the functional C-terminal domain of YT521-B was confirmed by pull-down assays and biomolecular interaction analysis (BIAcore). Inhibition by emerin of YT521-B-dependent splice site selection in vivo suggests that the interaction is physiologically significant. A 'bipartite' binding site for YT521-B in emerin was identified using alanine substitution or disease-associated mutations in emerin. The transcription factor GCL (germ cell-less) has previously been shown to bind to the same site. The results are consistent with an emerging view that lamins and lamina-associated proteins, like emerin, have a regulatory role, as well as a structural role in the nucleus. YT521-B joins a growing list of candidates for a role in a gene expression model of the pathogenesis of EDMD.


Asunto(s)
Proteínas de Drosophila , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Timopoyetinas/metabolismo , Sitios de Unión , Núcleo Celular/metabolismo , Cisteína Endopeptidasas/metabolismo , ADN Complementario/metabolismo , Biblioteca de Genes , Genes Reporteros , Humanos , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intercelular , Lamina Tipo A/química , Proteínas de la Membrana/química , Complejos Multienzimáticos/metabolismo , Distrofia Muscular de Emery-Dreifuss/metabolismo , Miocardio/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas Nucleares/metabolismo , Sistemas de Lectura Abierta , Pruebas de Precipitina , Complejo de la Endopetidasa Proteasomal , Unión Proteica , Estructura Terciaria de Proteína , ARN/metabolismo , Empalme del ARN , Factores de Empalme de ARN , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Timopoyetinas/química , Factores de Tiempo , Técnicas del Sistema de Dos Híbridos
17.
J Cell Biol ; 158(3): 475-85, 2002 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-12163470

RESUMEN

Barrier-to-autointegration factor (BAF) is a DNA-bridging protein, highly conserved in metazoans. BAF binds directly to LEM (LAP2, emerin, MAN1) domain nuclear membrane proteins, including LAP2 and emerin. We used site-directed mutagenesis and biochemical analysis to map functionally important residues in human BAF, including those required for direct binding to DNA or emerin. We also tested wild-type BAF and 25 point mutants for their effects on nuclear assembly in Xenopus egg extracts, which contain approximately 12 microM endogenous BAF dimers. Exogenous BAF caused two distinct effects: at low added concentrations, wild-type BAF enhanced chromatin decondensation and nuclear growth; at higher added concentrations, wild-type BAF completely blocked chromatin decondensation and nuclear growth. Mutants fell into four classes, including one that defines a novel functional surface on the BAF dimer. Our results suggest that BAF, unregulated, potently compresses chromatin structure, and that BAF interactions with both DNA and LEM proteins are critical for membrane recruitment and chromatin decondensation during nuclear assembly.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión al ADN/aislamiento & purificación , Proteínas de Unión al ADN/metabolismo , Células Eucariotas/metabolismo , Proteínas de la Membrana/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares , Timopoyetinas/metabolismo , Secuencia de Aminoácidos/genética , Animales , Células Cultivadas , Cromatina/genética , Clonación Molecular , ADN Complementario/análisis , Proteínas de Unión al ADN/genética , Dimerización , Células Eucariotas/ultraestructura , Femenino , Proteínas de la Membrana/genética , Microscopía Electrónica , Mutagénesis Sitio-Dirigida , Membrana Nuclear/ultraestructura , Oocitos , Fenotipo , Mutación Puntual/fisiología , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Timopoyetinas/genética , Xenopus laevis
18.
J Cell Biol ; 158(1): 53-62, 2002 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-12093788

RESUMEN

We tested the hypothesis that gp210, an integral membrane protein of nuclear pore complexes (NPCs), mediates nuclear pore formation. Gp210 has a large lumenal domain and small COOH-terminal tail exposed to the cytoplasm. We studied the exposed tail. We added recombinant tail polypeptides to Xenopus nuclear assembly extracts, or inhibited endogenous gp210 tails using anti-tail antibodies. Both strategies had no effect on the formation of fused flattened nuclear membranes, but blocked NPC assembly and nuclear growth. Inhibited nuclei accumulated gp210 and some nucleoporin p62, but failed to incorporate nup214/CAN, nup153, or nup98 and were defective for nuclear import of lamin B3. Scanning and transmission EM revealed a lack of "closely apposed" inner and outer membranes, and the accumulation of novel arrested structures including "mini-pores." We conclude that gp210 has early roles in nuclear pore formation, and that pore dilation is mediated by gp210 and its tail-binding partner(s). We propose that membrane fusion and pore dilation are coupled, acting as a mechanism to control nuclear pore size.


Asunto(s)
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas Nucleares/metabolismo , Secuencia de Aminoácidos , Animales , Membrana Celular/metabolismo , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Glutatión Transferasa/metabolismo , Immunoblotting , Proteínas de Filamentos Intermediarios/metabolismo , Lamina Tipo B , Fusión de Membrana , Microscopía Electrónica , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Datos de Secuencia Molecular , Proteínas de Complejo Poro Nuclear/metabolismo , Estructura Terciaria de Proteína , Radiometría , Proteínas Recombinantes de Fusión/metabolismo , Trombina/metabolismo , Xenopus , Proteínas de Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA