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










Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 295(13): 4194-4211, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32071079

RESUMO

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.


Assuntos
Autoantígenos/genética , Hidrolases de Éster Carboxílico/genética , Proteínas de Ligação a DNA/genética , Chaperonas de Histonas/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Proteína Fosfatase 2/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Neoplasias/genética , Neoplasias/patologia , Neoplasias/terapia , Lâmina Nuclear/efeitos dos fármacos , Lâmina Nuclear/genética , Fosfoproteínas/antagonistas & inibidores , Fosfoproteínas/genética , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 2/genética , Proteoma/efeitos dos fármacos , Proto-Oncogene Mas , RNA Interferente Pequeno/genética , Biologia de Sistemas
2.
Nucleus ; 10(1): 7-20, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-30663495

RESUMO

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.


Assuntos
Cromatina/metabolismo , Lamina Tipo A/metabolismo , Lamina Tipo B/metabolismo , Lâmina Nuclear/metabolismo , Cromatina/efeitos dos fármacos , Ciclosporina/farmacologia , Células Hep G2 , Humanos , Hibridização in Situ Fluorescente , Lamina Tipo A/antagonistas & inibidores , Lamina Tipo B/antagonistas & inibidores , Modelos Genéticos , Lâmina Nuclear/efeitos dos fármacos , Células Tumorais Cultivadas
3.
Aging Cell ; 18(1): e12851, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30565836

RESUMO

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.


Assuntos
Cromatina/metabolismo , Dano ao DNA , Lâmina Nuclear/metabolismo , Transdução de Sinais , Proteína ran de Ligação ao GTP/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Azepinas/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Histonas/metabolismo , Humanos , Interfase/efeitos dos fármacos , Lamina Tipo A/metabolismo , Lisina/metabolismo , Metilação/efeitos dos fármacos , Lâmina Nuclear/efeitos dos fármacos , Progéria/patologia , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos
4.
J Virol ; 88(18): 10982-5, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24965476

RESUMO

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.


Assuntos
Núcleo Celular/virologia , Infecções por Citomegalovirus/enzimologia , Citomegalovirus/enzimologia , Lâmina Nuclear/virologia , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Liberação de Vírus/efeitos dos fármacos , Capsídeo/metabolismo , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/genética , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/virologia , Humanos , Lâmina Nuclear/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteína Quinase C/metabolismo , Montagem de Vírus/efeitos dos fármacos
5.
Virology ; 406(1): 127-37, 2010 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-20674954

RESUMO

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.


Assuntos
Herpesvirus Humano 1/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Lâmina Nuclear/virologia , Proteína Quinase C/fisiologia , Liberação de Vírus/fisiologia , Animais , Sequência de Bases , Capsídeo/efeitos dos fármacos , Capsídeo/fisiologia , Capsídeo/ultraestrutura , Linhagem Celular , Chlorocebus aethiops , Primers do DNA/genética , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/ultraestrutura , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Microscopia Eletrônica de Transmissão , Modelos Biológicos , Lâmina Nuclear/efeitos dos fármacos , Lâmina Nuclear/enzimologia , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Células Vero , Montagem de Vírus/efeitos dos fármacos , Montagem de Vírus/fisiologia , Liberação de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Replicação Viral/fisiologia
6.
Nucleic Acids Res ; 37(7): 2238-48, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19237397

RESUMO

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.


Assuntos
Núcleo Celular/genética , Replicação do DNA , Animais , Cromatina/metabolismo , Replicação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Etídio/farmacologia , Substâncias Intercalantes/farmacologia , Membrana Nuclear/efeitos dos fármacos , Lâmina Nuclear/efeitos dos fármacos , Xenopus
7.
Exp Cell Res ; 314(6): 1392-405, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18294630

RESUMO

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.


Assuntos
Diferenciação Celular , Lamina Tipo A/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Distrofia Muscular de Emery-Dreifuss/enzimologia , Mioblastos/citologia , Mioblastos/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Antígenos CD1/metabolismo , Complexo CD3/metabolismo , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Proteínas de Fluorescência Verde/metabolismo , Fator de Crescimento Insulin-Like II/farmacologia , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Distrofia Muscular de Emery-Dreifuss/patologia , Proteínas Mutantes/metabolismo , Mutação/genética , Mioblastos/efeitos dos fármacos , Miogenina/metabolismo , Lâmina Nuclear/efeitos dos fármacos , Lâmina Nuclear/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteína do Retinoblastoma/metabolismo
8.
Exp Cell Res ; 314(3): 453-62, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18093584

RESUMO

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.


Assuntos
Núcleo Celular/efeitos dos fármacos , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Farnesiltranstransferase/antagonistas & inibidores , Heterocromatina/efeitos dos fármacos , Proteínas Nucleares/efeitos dos fármacos , Precursores de Proteínas/efeitos dos fármacos , Prenilação de Proteína/efeitos dos fármacos , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Adulto , Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Células Cultivadas , Montagem e Desmontagem da Cromatina/genética , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Decitabina , Inibidores Enzimáticos/farmacologia , Farnesiltranstransferase/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Heterocromatina/genética , Heterocromatina/ultraestrutura , Humanos , Lamina Tipo A/efeitos dos fármacos , Lamina Tipo A/metabolismo , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Metionina/análogos & derivados , Metionina/farmacologia , Lâmina Nuclear/efeitos dos fármacos , Lâmina Nuclear/metabolismo , Lâmina Nuclear/ultraestrutura , Proteínas Nucleares/metabolismo , Precursores de Proteínas/metabolismo , Prenilação de Proteína/fisiologia
9.
J Cell Sci ; 120(Pt 9): 1673-80, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17430977

RESUMO

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.


Assuntos
Núcleo Celular/metabolismo , Proteínas Nucleares/análise , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/ultraestrutura , Estruturas do Núcleo Celular/efeitos dos fármacos , Estruturas do Núcleo Celular/metabolismo , Estruturas do Núcleo Celular/ultraestrutura , Cromatina/efeitos dos fármacos , Cromatina/metabolismo , Cromatina/ultraestrutura , Dextranos/farmacologia , Digitonina/farmacologia , Células HeLa , Histonas/análise , Humanos , Soluções Hipertônicas/farmacologia , Lamina Tipo A/análise , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Lâmina Nuclear/efeitos dos fármacos , Lâmina Nuclear/metabolismo , Lâmina Nuclear/ultraestrutura , Pressão Osmótica , Ribonucleoproteínas/análise , Fatores de Processamento de Serina-Arginina , Cloreto de Sódio/farmacologia , Sorbitol/farmacologia , Sacarose/farmacologia
10.
J Cell Biol ; 168(2): 245-55, 2005 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-15657395

RESUMO

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.


Assuntos
Actinas/metabolismo , Apoptose/fisiologia , Núcleo Celular/metabolismo , Laminas/metabolismo , Miosinas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Amidas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Inibidores de Caspase , Caspases/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/ultraestrutura , Cicloeximida/farmacologia , Citocalasina D/farmacologia , Proteínas do Citoesqueleto , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/ultraestrutura , Peptídeos e Proteínas de Sinalização Intracelular , Laminas/genética , Quinases Lim , Camundongos , Microscopia Eletrônica de Transmissão , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Mutação/fisiologia , Cadeias Leves de Miosina/genética , Cadeias Leves de Miosina/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Miosinas/antagonistas & inibidores , Células NIH 3T3 , Nocodazol/farmacologia , Lâmina Nuclear/efeitos dos fármacos , Lâmina Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Piridinas/farmacologia , Transfecção , Fator de Necrose Tumoral alfa/farmacologia , Quinases Associadas a rho
11.
J Cell Biol ; 167(6): 1051-62, 2004 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-15611332

RESUMO

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.


Assuntos
Metabolismo Energético/fisiologia , Proteínas de Membrana/fisiologia , Poro Nuclear/fisiologia , Proteínas Nucleares/fisiologia , Trifosfato de Adenosina/farmacologia , Retículo Endoplasmático/fisiologia , Células HeLa , Humanos , Proteínas de Membrana/efeitos dos fármacos , Lâmina Nuclear/efeitos dos fármacos , Lâmina Nuclear/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Sirolimo/farmacologia , Temperatura , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...