Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 295(13): 4194-4211, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32071079

RESUMEN

Protein phosphatase 2A (PP2A) critically regulates cell signaling and is a human tumor suppressor. PP2A complexes are modulated by proteins such as cancerous inhibitor of protein phosphatase 2A (CIP2A), protein phosphatase methylesterase 1 (PME-1), and SET nuclear proto-oncogene (SET) that often are deregulated in cancers. However, how they impact cellular phosphorylation and how redundant they are in cellular regulation is poorly understood. Here, we conducted a systematic phosphoproteomics screen for phosphotargets modulated by siRNA-mediated depletion of CIP2A, PME-1, and SET (to reactivate PP2A) or the scaffolding A-subunit of PP2A (PPP2R1A) (to inhibit PP2A) in HeLa cells. We identified PP2A-modulated targets in diverse cellular pathways, including kinase signaling, cytoskeleton, RNA splicing, DNA repair, and nuclear lamina. The results indicate nonredundancy among CIP2A, PME-1, and SET in phosphotarget regulation. Notably, PP2A inhibition or reactivation affected largely distinct phosphopeptides, introducing a concept of nonoverlapping phosphatase inhibition- and activation-responsive sites (PIRS and PARS, respectively). This phenomenon is explained by the PPP2R1A inhibition impacting primarily dephosphorylated threonines, whereas PP2A reactivation results in dephosphorylation of clustered and acidophilic sites. Using comprehensive drug-sensitivity screening in PP2A-modulated cells to evaluate the functional impact of PP2A across diverse cellular pathways targeted by these drugs, we found that consistent with global phosphoproteome effects, PP2A modulations broadly affect responses to more than 200 drugs inhibiting a broad spectrum of cancer-relevant targets. These findings advance our understanding of the phosphoproteins, pharmacological responses, and cellular processes regulated by PP2A modulation and may enable the development of combination therapies.


Asunto(s)
Autoantígenos/genética , Hidrolasas de Éster Carboxílico/genética , Proteínas de Unión al ADN/genética , Chaperonas de Histonas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Proteína Fosfatasa 2/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/química , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Lámina Nuclear/efectos de los fármacos , Lámina Nuclear/genética , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2/genética , Proteoma/efectos de los fármacos , Proto-Oncogenes Mas , ARN Interferente Pequeño/genética , Biología de Sistemas
2.
Nucleus ; 10(1): 7-20, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30663495

RESUMEN

Immunosuppressive drugs such as cyclosporin A (CsA) can elicit hepatotoxicity by affecting gene expression. Here, we address the link between CsA and large-scale chromatin organization in HepG2 hepatocarcinoma cells. We show the existence of lamina-associated domains (LADs) interacting with lamin A, lamin B, or both. These 'A-B', 'A-only' and 'B-only' LADs display distinct fates after CsA treatment: A-B LADs remain constitutive or lose A, A-only LADs mainly lose A or switch to B, and B-only LADs remain B-only or acquire A. LAD rearrangement is overall uncoupled from changes in gene expression. Three-dimensional (3D) genome modeling predicts changes in radial positioning of LADs as LADs switch identities, which are corroborated by fluorescence in situ hybridization. Our results reveal interplay between A- and B-type lamins on radial locus positioning, suggesting complementary contributions to large-scale genome architecture. The data also unveil a hitherto unsuspected impact of cytotoxic drugs on genome conformation.Abbreviations: ChIP-seq: chromatin immunoprecipitation sequencing; CsA: cyclosporin A; FISH; fluorescence in situ hybridization; ICMT: isoprenylcysteine methyltransferase; LAD: lamina-associated domain; TAD: topologically-associated domain.


Asunto(s)
Cromatina/metabolismo , Lamina Tipo A/metabolismo , Lamina Tipo B/metabolismo , Lámina Nuclear/metabolismo , Cromatina/efectos de los fármacos , Ciclosporina/farmacología , Células Hep G2 , Humanos , Hibridación Fluorescente in Situ , Lamina Tipo A/antagonistas & inhibidores , Lamina Tipo B/antagonistas & inhibidores , Modelos Genéticos , Lámina Nuclear/efectos de los fármacos , Células Tumorales Cultivadas
3.
Aging Cell ; 18(1): e12851, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30565836

RESUMEN

The Ran GTPase regulates nuclear import and export by controlling the assembly state of transport complexes. This involves the direct action of RanGTP, which is generated in the nucleus by the chromatin-associated nucleotide exchange factor, RCC1. Ran interactions with RCC1 contribute to formation of a nuclear:cytoplasmic (N:C) Ran protein gradient in interphase cells. In previous work, we showed that the Ran protein gradient is disrupted in fibroblasts from Hutchinson-Gilford progeria syndrome (HGPS) patients. The Ran gradient disruption in these cells is caused by nuclear membrane association of a mutant form of Lamin A, which induces a global reduction in heterochromatin marked with Histone H3K9me3 and Histone H3K27me3. Here, we have tested the hypothesis that heterochromatin controls the Ran gradient. Chemical inhibition and depletion of the histone methyltransferases (HMTs) G9a and GLP in normal human fibroblasts reduced heterochromatin levels and caused disruption of the Ran gradient, comparable to that observed previously in HGPS fibroblasts. HMT inhibition caused a defect in nuclear localization of TPR, a high molecular weight protein that, owing to its large size, displays a Ran-dependent import defect in HGPS. We reasoned that pathways dependent on nuclear import of large proteins might be compromised in HGPS. We found that nuclear import of ATM requires the Ran gradient, and disruption of the Ran gradient in HGPS causes a defect in generating nuclear γ-H2AX in response to ionizing radiation. Our data suggest a lamina-chromatin-Ran axis is important for nuclear transport regulation and contributes to the DNA damage response.


Asunto(s)
Cromatina/metabolismo , Daño del ADN , Lámina Nuclear/metabolismo , Transducción de Señal , Proteína de Unión al GTP ran/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Azepinas/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Histonas/metabolismo , Humanos , Interfase/efectos de los fármacos , Lamina Tipo A/metabolismo , Lisina/metabolismo , Metilación/efectos de los fármacos , Lámina Nuclear/efectos de los fármacos , Progeria/patología , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos
4.
J Virol ; 88(18): 10982-5, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24965476

RESUMEN

Human cytomegalovirus (HCMV) kinase UL97 is required for efficient nuclear lamina disruption during nuclear egress. However, cellular protein kinase C (PKC) has been implicated in this process in other systems. Comparing the effects of UL97 and cellular kinase inhibitors on HCMV nuclear egress confirms a role for UL97 in lamina disruption and nuclear egress. A pan-PKC inhibitor did not affect lamina disruption but did reduce the number of cytoplasmic capsids more than the number of nuclear capsids.


Asunto(s)
Núcleo Celular/virología , Infecciones por Citomegalovirus/enzimología , Citomegalovirus/enzimología , Lámina Nuclear/virología , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Liberación del Virus/efectos de los fármacos , Cápside/metabolismo , Línea Celular , Núcleo Celular/efectos de los fármacos , Citomegalovirus/efectos de los fármacos , Citomegalovirus/genética , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/virología , Humanos , Lámina Nuclear/efectos de los fármacos , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteína Quinasa C/metabolismo , Ensamble de Virus/efectos de los fármacos
5.
Virology ; 406(1): 127-37, 2010 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-20674954

RESUMEN

The nuclear lamina is thought to be a steric barrier to the herpesvirus capsid. Disruption of the lamina accompanied by phosphorylation of lamina proteins is a conserved feature of herpesvirus infection. In HSV-1-infected cells, protein kinase C (PKC) alpha and delta isoforms are recruited to the nuclear membrane and PKC delta has been implicated in phosphorylation of emerin and lamin B. We tested two critical hypotheses about the mechanism and significance of lamina disruption. First, we show that chemical inhibition of all PKC isoforms reduced viral growth five-fold and inhibited capsid egress from the nucleus. However, specific inhibition of either conventional PKCs or PKC delta does not inhibit viral growth. Second, we show hyperphosphorylation of emerin by viral and cellular kinases is required for its disassociation from the lamina. These data support hypothesis that phosphorylation of lamina components mediates lamina disruption during HSV nuclear egress.


Asunto(s)
Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno/fisiología , Lámina Nuclear/virología , Proteína Quinasa C/fisiología , Liberación del Virus/fisiología , Animales , Secuencia de Bases , Cápside/efectos de los fármacos , Cápside/fisiología , Cápside/ultraestructura , Línea Celular , Chlorocebus aethiops , Cartilla de ADN/genética , Herpesvirus Humano 1/efectos de los fármacos , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/ultraestructura , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Microscopía Electrónica de Transmisión , Modelos Biológicos , Lámina Nuclear/efectos de los fármacos , Lámina Nuclear/enzimología , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Células Vero , Ensamble de Virus/efectos de los fármacos , Ensamble de Virus/fisiología , Liberación del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología
6.
Nucleic Acids Res ; 37(7): 2238-48, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19237397

RESUMEN

In this article, we study how intercalation-induced changes in chromatin and DNA topology affect chromosomal DNA replication using Xenopus egg extracts. Unexpectedly, intercalation by ethidium or doxorubicin prevents formation of a functional nucleus: although nucleosome formation occurs, DNA decondensation is arrested, membranous vesicles accumulate around DNA but do not fuse to form a nuclear membrane, active transport is abolished and lamins are found on chromatin, but do not assemble into a lamina. DNA replication is inhibited at the stage of initiation complex activation, as shown by molecular combing of DNA and by the absence of checkpoint activation. Replication of single-stranded DNA is not prevented. Surprisingly, in spite of the absence of nuclear function, DNA-replication proteins of pre-replication and initiation complexes are loaded onto chromatin. This is a general phenomenon as initiation complexes could also be seen without ethidium in membrane-depleted extracts which do not form nuclei. These results suggest that DNA or chromatin topology is required for generation of a functional nucleus, and activation, but not formation, of initiation complexes.


Asunto(s)
Núcleo Celular/genética , Replicación del ADN , Animales , Cromatina/metabolismo , Replicación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Etidio/farmacología , Sustancias Intercalantes/farmacología , Membrana Nuclear/efectos de los fármacos , Lámina Nuclear/efectos de los fármacos , Xenopus
7.
Exp Cell Res ; 314(6): 1392-405, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18294630

RESUMEN

Mutation R453W in A-type lamins, that are major nuclear envelope proteins, generates Emery-Dreifuss muscular dystrophy. We previously showed that mouse myoblasts expressing R453W-lamin A incompletely exit the cell cycle and differentiate into myocytes with a low level of multinucleation. Here we attempted to improve differentiation by treating these cells with a mixture of PD98059, an extracellular-regulated kinase (ERK) kinase (also known as mitogen-activated kinase, MEK) inhibitor, and insulin-like growth factor-II, an activator of phosphoinositide 3-kinase. We show that mouse myoblasts expressing R453W-lamin A were sensitive to the drug treatment as shown by (i) an increase in multinucleation, (ii) downregulation of proliferation markers (cyclin D1, hyperphosphorylated Rb), (iii) upregulation of myogenin, and (iv) sustained activation of p21 and cyclin D3. However, nuclear matrix anchorage of p21 and cyclin D3 in a complex with hypophosphorylated Rb that is critical to trigger cell cycle arrest and myogenin induction was deficient and incompletely restored by drug treatment. As the turn-over of R453W-lamin A at the nuclear envelope was greatly enhanced, we propose that R453W-lamin A impairs the capacity of the nuclear lamina to serve as scaffold for substrates of the MEK-ERK pathway and for MyoD-induced proteins that play a role in the differentiation process.


Asunto(s)
Diferenciación Celular , Lamina Tipo A/metabolismo , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Distrofia Muscular de Emery-Dreifuss/enzimología , Mioblastos/citología , Mioblastos/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Antígenos CD1/metabolismo , Complejo CD3/metabolismo , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Proteínas Fluorescentes Verdes/metabolismo , Factor II del Crecimiento Similar a la Insulina/farmacología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Distrofia Muscular de Emery-Dreifuss/patología , Proteínas Mutantes/metabolismo , Mutación/genética , Mioblastos/efectos de los fármacos , Miogenina/metabolismo , Lámina Nuclear/efectos de los fármacos , Lámina Nuclear/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteína de Retinoblastoma/metabolismo
8.
Exp Cell Res ; 314(3): 453-62, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18093584

RESUMEN

Increasing interest in drugs acting on prelamin A has derived from the finding of prelamin A involvement in severe laminopathies. Amelioration of the nuclear morphology by inhibitors of prelamin A farnesylation has been widely reported in progeroid laminopathies. We investigated the effects on chromatin organization of two drugs inhibiting prelamin A processing by an ultrastructural and biochemical approach. The farnesyltransferase inhibitor FTI-277 and the non-peptidomimetic drug N-acetyl-S-farnesyl-l-cysteine methylester (AFCMe) were administered to cultured control human fibroblasts for 6 or 18 h. FTI-277 interferes with protein farnesylation causing accumulation of non-farnesylated prelamin A, while AFCMe impairs the last cleavage of the lamin A precursor and is expected to accumulate farnesylated prelamin A. FTI-277 caused redistribution of heterochromatin domains at the nuclear interior, while AFCMe caused loss of heterochromatin domains, increase of nuclear size and nuclear lamina thickening. At the biochemical level, heterochromatin-associated proteins and LAP2 alpha were clustered at the nuclear interior following FTI-277 treatment, while they were unevenly distributed or absent in AFCMe-treated nuclei. The reported effects show that chromatin is an immediate target of FTI-277 and AFCMe and that dramatic remodeling of chromatin domains occurs following treatment with the drugs. These effects appear to depend, at least in part, on the accumulation of prelamin A forms, since impairment of prelamin A accumulation, here obtained by 5-azadeoxycytidine treatment, abolishes the chromatin effects. These results may be used to evaluate downstream effects of FTIs or other prelamin A inhibitors potentially useful for the therapy of laminopathies.


Asunto(s)
Núcleo Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Farnesiltransferasa/antagonistas & inhibidores , Heterocromatina/efectos de los fármacos , Proteínas Nucleares/efectos de los fármacos , Precursores de Proteínas/efectos de los fármacos , Prenilación de Proteína/efectos de los fármacos , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Adulto , Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Azacitidina/farmacología , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Células Cultivadas , Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Decitabina , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Heterocromatina/genética , Heterocromatina/ultraestructura , Humanos , Lamina Tipo A/efectos de los fármacos , Lamina Tipo A/metabolismo , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Metionina/análogos & derivados , Metionina/farmacología , Lámina Nuclear/efectos de los fármacos , Lámina Nuclear/metabolismo , Lámina Nuclear/ultraestructura , Proteínas Nucleares/metabolismo , Precursores de Proteínas/metabolismo , Prenilación de Proteína/fisiología
9.
J Cell Sci ; 120(Pt 9): 1673-80, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17430977

RESUMEN

Many compounds in the cell nucleus are structurally organized. To assess the influence of structural organization on nuclear function, we investigated the physical mechanisms of structure formation by using molecular crowding as a parameter for nuclear integrity. Molecular crowding promotes compaction of macromolecular compounds depending on their size and shape without the need for site-specific interactions. HeLa and MCF7 cells were incubated with hypertonic medium to increase crowding of their macromolecular content as a result of the osmotic loss of water. Supplementation of sucrose, sorbitol or NaCl to the growth medium shifted nuclear organization, observed by fluorescence and electron microscopy, towards compaction of chromatin and segregation of other nuclear compounds. With increasing hypertonic load and incubation time, this nuclear re-organization proceeded gradually, irrespective of the substances used, and reversibly relaxed to a regular phenotype upon re-incubation of cells in isotonic growth medium. Gradual and reversible re-organization are major features of controlled de-mixing by molecular crowding. Of fundamental importance for nuclear function, we discuss how macromolecular crowding could account for the stabilization of processes that involve large, macromolecular machines.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas Nucleares/análisis , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Estructuras del Núcleo Celular/efectos de los fármacos , Estructuras del Núcleo Celular/metabolismo , Estructuras del Núcleo Celular/ultraestructura , Cromatina/efectos de los fármacos , Cromatina/metabolismo , Cromatina/ultraestructura , Dextranos/farmacología , Digitonina/farmacología , Células HeLa , Histonas/análisis , Humanos , Soluciones Hipertónicas/farmacología , Lamina Tipo A/análisis , Microscopía Confocal , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Lámina Nuclear/efectos de los fármacos , Lámina Nuclear/metabolismo , Lámina Nuclear/ultraestructura , Presión Osmótica , Ribonucleoproteínas/análisis , Factores de Empalme Serina-Arginina , Cloruro de Sodio/farmacología , Sorbitol/farmacología , Sacarosa/farmacología
10.
J Cell Biol ; 168(2): 245-55, 2005 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-15657395

RESUMEN

Membrane blebbing during the apoptotic execution phase results from caspase-mediated cleavage and activation of ROCK I. Here, we show that ROCK activity, myosin light chain (MLC) phosphorylation, MLC ATPase activity, and an intact actin cytoskeleton, but not microtubular cytoskeleton, are required for disruption of nuclear integrity during apoptosis. Inhibition of ROCK or MLC ATPase activity, which protect apoptotic nuclear integrity, does not affect caspase-mediated degradation of nuclear proteins such as lamins A, B1, or C. The conditional activation of ROCK I was sufficient to tear apart nuclei in lamin A/C null fibroblasts, but not in wild-type fibroblasts. Thus, apoptotic nuclear disintegration requires actin-myosin contractile force and lamin proteolysis, making apoptosis analogous to, but distinct from, mitosis where nuclear disintegration results from microtubule-based forces and from lamin phosphorylation and depolymerization.


Asunto(s)
Actinas/metabolismo , Apoptosis/fisiología , Núcleo Celular/metabolismo , Laminas/metabolismo , Miosinas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Amidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Caspasas/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Cicloheximida/farmacología , Citocalasina D/farmacología , Proteínas del Citoesqueleto , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Inhibidores Enzimáticos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/ultraestructura , Péptidos y Proteínas de Señalización Intracelular , Laminas/genética , Quinasas Lim , Ratones , Microscopía Electrónica de Transmisión , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Mutación/fisiología , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Miosinas/antagonistas & inhibidores , Células 3T3 NIH , Nocodazol/farmacología , Lámina Nuclear/efectos de los fármacos , Lámina Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Piridinas/farmacología , Transfección , Factor de Necrosis Tumoral alfa/farmacología , Quinasas Asociadas a rho
11.
J Cell Biol ; 167(6): 1051-62, 2004 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-15611332

RESUMEN

Resident integral proteins of the inner nuclear membrane (INM) are synthesized as membrane-integrated proteins on the peripheral endoplasmic reticulum (ER) and are transported to the INM throughout interphase using an unknown trafficking mechanism. To study this transport, we developed a live cell assay that measures the movement of transmembrane reporters from the ER to the INM by rapamycin-mediated trapping at the nuclear lamina. Reporter constructs with small (<30 kD) cytosolic and lumenal domains rapidly accumulated at the INM. However, increasing the size of either domain by 47 kD strongly inhibited movement. Reduced temperature and ATP depletion also inhibited movement, which is characteristic of membrane fusion mechanisms, but pharmacological inhibition of vesicular trafficking had no effect. Because reporter accumulation at the INM was inhibited by antibodies to the nuclear pore membrane protein gp210, our results support a model wherein transport of integral proteins to the INM involves lateral diffusion in the lipid bilayer around the nuclear pore membrane, coupled with active restructuring of the nuclear pore complex.


Asunto(s)
Metabolismo Energético/fisiología , Proteínas de la Membrana/fisiología , Poro Nuclear/fisiología , Proteínas Nucleares/fisiología , Adenosina Trifosfato/farmacología , Retículo Endoplásmico/fisiología , Células HeLa , Humanos , Proteínas de la Membrana/efectos de los fármacos , Lámina Nuclear/efectos de los fármacos , Lámina Nuclear/fisiología , Proteínas de Complejo Poro Nuclear/fisiología , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Sirolimus/farmacología , Temperatura , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...