Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
STAR Protoc ; 2(3): 100649, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34278335

RESUMO

The cyclic GMP-AMP synthase (cGAS) is the principal DNA sensor, which binds DNA and triggers the type I interferon production. We used ISD45 or inactivated Vaccinia Virus (VACV) to stimulate cGAS and monitored cellular localization by immunofluorescence microscopy, Operetta high-content screening, and cytoplasmic/nuclear fractionation. LocNES server was used to predict cGAS nuclear export signal (NES) sequence and characterized the function by mutagenesis. This protocol provides a prototype of cGAS subcellular distribution or the identification of NES in other proteins. For complete details on the use and execution of this protocol, please refer to Sun et al. Sun et al. (2021).


Assuntos
Microscopia de Fluorescência/métodos , Biologia Molecular/métodos , Sinais de Exportação Nuclear/fisiologia , Nucleotidiltransferases/metabolismo , Animais , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Mamíferos , Mutagênese , Sinais de Exportação Nuclear/genética , Nucleotidiltransferases/genética , Vaccinia virus/genética
3.
FEBS Lett ; 594(10): 1596-1607, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32052428

RESUMO

Replication protein A (RPA), a heterotrimeric complex, is the major single-stranded DNA binding protein in eukaryotes. Recently, we characterized RPA from Trypanosoma cruzi, showing that it is involved in DNA replication and DNA damage response in this organism. Better efficiency in differentiation from epimastigote to metacyclic trypomastigote forms was observed in TcRPA-2 subunit heterozygous knockout cells, suggesting that RPA is involved in this process. Here, we show that RPA cellular localization changes during the T. cruzi life cycle, with RPA being detected only in the cytoplasm of the metacyclic and bloodstream trypomastigotes. We also identify a nuclear export signal (NES) in the trypanosomatid RPA-2 subunit. Mutations in the negatively charged residues of RPA-2 NES impair the differentiation process, suggesting that RPA exportation affects parasite differentiation into infective forms.


Assuntos
Núcleo Celular/metabolismo , Estágios do Ciclo de Vida , Morfogênese , Proteína de Replicação A/metabolismo , Trypanosoma cruzi/crescimento & desenvolvimento , Trypanosoma cruzi/metabolismo , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Doença de Chagas/sangue , Doença de Chagas/parasitologia , Simulação por Computador , Citoplasma/metabolismo , Morfogênese/genética , Sinais de Exportação Nuclear/genética , Sinais de Exportação Nuclear/fisiologia , Proteína de Replicação A/genética , Trypanosoma cruzi/citologia
4.
Cells ; 8(10)2019 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-31614958

RESUMO

Nuclear-cytoplasmic shuttling is a highly regulated and complex process, which involves both proteins and nucleic acids. Changes in cellular compartmentalization of various proteins, including oncogenes and tumor suppressors, affect cellular behavior, promoting or inhibiting proliferation, apoptosis and sensitivity to therapies. In this review, we will recapitulate the role of various shuttling components in Chronic Myeloid Leukemia and we will provide insights on the potential role of shuttling proteins as therapeutic targets.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/fisiopatologia , Apoptose , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Citosol/metabolismo , Humanos , Hidrazinas/farmacologia , Carioferinas/metabolismo , Sinais de Exportação Nuclear/fisiologia , Sinais de Localização Nuclear/metabolismo , Sinais de Localização Nuclear/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Triazóis/farmacologia , Proteína Exportina 1
5.
Sci Rep ; 9(1): 6627, 2019 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-31036839

RESUMO

Nuclear export signal (NES) motifs function as essential regulators of the subcellular location of proteins by interacting with the major nuclear exporter protein, CRM1. Prediction of NES is of great interest in many aspects of research including cancer, but currently available methods, which are mostly based on the sequence-based approaches, have been suffered from high false positive rates since the NES consensus patterns are quite commonly observed in protein sequences. Therefore, finding a feature that can distinguish real NES motifs from false positives is desired to improve the prediction power, but it is quite challenging when only using the sequence. Here, we provide a comprehensive table for the validated cargo proteins, containing the location of the NES consensus patterns with the disordered propensity plots, known protein domain information, and the predicted secondary structures. It could be useful for determining the most plausible NES region in the context of the whole protein sequence and suggests possibilities for some non-binders of the annotated regions. In addition, using the currently available crystal structures of CRM1 bound to various classes of NES peptides, we adopted, for the first time, the structure-based prediction of the NES motifs bound to the CRM1's binding groove. Combining sequence-based and structure-based predictions, we suggest a novel and more straight-forward approach to identify CRM1-binding NES sequences by analysis of their structural prerequisites and energetic evaluation of the stability at the CRM1's binding site.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Núcleo Celular/metabolismo , Sinais de Exportação Nuclear/fisiologia , Transporte Ativo do Núcleo Celular/genética , Sítios de Ligação , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Sinais de Exportação Nuclear/genética , Ligação Proteica , Estrutura Secundária de Proteína , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo
6.
Beijing Da Xue Xue Bao Yi Xue Ban ; 49(4): 569-574, 2017 08 18.
Artigo em Chinês | MEDLINE | ID: mdl-28816267

RESUMO

OBJECTIVE: To investigate the mechanisms of nuclear export signal of androgen receptor (NESAR) in the regulation of androgen receptor (AR) protein expression and stability in prostate cancer. METHODS: The green fluorescent protein fusion protein expression vectors pEGFP-AR(1-918aa), pEGFP-NESAR (743-817aa), pEGFP-NAR (1-665aa) and pEGFP-NAR-NESAR, and lysine mutants of NESAR pEGFP-NESAR K776R, pEGFP-NESAR K807R and pEGFP-NESAR K776R/K807R, were transiently transfected into prostate cancer cell line PC3. Fluorescence microscopy, Western blot and immunoprecipitation were used to detect NESAR regulation of androgen receptor stability. RESULTS: Under the fluorescence microscope, NESAR-containing fusion proteins were cytoplasmic localization, and their fluorescence intensities were much weaker than those without NESAR. The expression levels of NESAR-containing fusion proteins were significantly lower than those without NESAR. The half-lives of GFP-NESAR and GFP-NAR-NESAR were less than 6 h, while the expression of GFP and GFP-NAR was relatively stable and the half-life was more than 24 h in the presence of cycloheximide. The expression levels of GFP-NESAR were significantly increased by proteasome inhibitor MG132 treatment in a dose-dependent manner; in contrast, MG132 did not show any significant effect on the protein levels of GFP. When new protein synthesis was blocked, MG132 could also prevent the degradation of GFP-NESAR in the transfected cells in the presence of cycloheximide, while it had no significant effect on GFP protein stability in the parallel experiment. GFP immunoprecipitation showed that the ubiquitination level of GFP-NESAR fusion protein was significantly higher than that of the GFP control. The mutations of lysine sites K776 and K807 in NESAR significantly reduced the level of ubiquitination, and showed increased protein stability, indicating that they were the key amino acid residues of NESAR ubiquitination. CONCLUSION: NESAR was unstable and decreased the stability of its fusion proteins. NESAR was the target of polyubiquitination and mediated the degradation of its fusion proteins through the ubiquitin-proteasome pathway in prostate cancer cells. Our research provides a new way to regulate the level and/or activity of AR proteins, thus helping us understand the molecular mechanisms of AR degradation and strict control of AR in the progression to castration-resistance.


Assuntos
Sinais de Exportação Nuclear , Neoplasias da Próstata , Receptores Androgênicos , Androgênios , Linhagem Celular Tumoral , Humanos , Masculino , Sinais de Exportação Nuclear/fisiologia , Neoplasias da Próstata/metabolismo , Receptores Androgênicos/fisiologia
7.
J Vis Exp ; (123)2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28570551

RESUMO

Nucleocytoplasmic transport refers to the import and export of large molecules from the cell nucleus. Recently, a number of studies have shown a connection between amyotrophic lateral sclerosis (ALS) and impairments in the nucleocytoplasmic pathway. ALS is a neurodegenerative disease affecting the motor neurons and resulting in paralysis and ultimately in death, within 2-5 years on average. Most cases of ALS are sporadic, lacking any apparent genetic linkage, but 10% are inherited in a dominant manner. Recently, hexanucleotide repeat expansions (HREs) in the chromosome 9 open reading frame 72 (C9orf72) gene were identified as a genetic cause of ALS and frontotemporal dementia (FTD). Importantly, different groups have recently proposed that these mutants affect nucleocytoplasmic transport. These studies have mostly shown the final outcome and manifestations caused by HREs on nucleocytoplasmic transport, but they do not demonstrate nuclear transport dysfunction in real time. As a result, only severe nucleocytoplasmic transport deficiency can be determined, mostly due to high overexpression or exogenous protein insertion. This protocol describes a new and very sensitive assay to evaluate and quantify nucleocytoplasmic transport dysfunction in real time. The rate of import of a NLS-NES-GFP protein (shuttle-GFP) can be quantified in real time using fluorescent microscopy. This is performed by using an exportin inhibitor, thus allowing the shuttle GFP only to enter the nucleus. To validate the assay, the C9orf72 HRE translated dipeptide repeats, poly(GR) and poly(PR), which have been previously shown to disrupt nucleocytoplasmic transport, were used. Using the described assay, a 50% decrease in the nuclear import rate was observed compared to the control. Using this system, minute changes in nucleocytoplasmic transport can be examined and the ability of different factors to rescue (even partially) a nucleocytoplasmic transport defect can be determined.


Assuntos
Proteína C9orf72/metabolismo , Neurônios Motores/metabolismo , Transporte Ativo do Núcleo Celular , Esclerose Lateral Amiotrófica/diagnóstico , Esclerose Lateral Amiotrófica/metabolismo , Animais , Proteína C9orf72/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Dipeptídeos/metabolismo , Demência Frontotemporal/diagnóstico , Demência Frontotemporal/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Sinais de Exportação Nuclear/fisiologia , Sinais de Localização Nuclear/metabolismo
8.
Mol Endocrinol ; 29(12): 1731-42, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26371515

RESUMO

Androgen receptor (AR) plays a pivotal role in the development of primary as well as advanced castration-resistant prostate cancer. Previous work in our lab identified a novel nuclear export signal (NES) (NES(AR)) in AR ligand-binding domain essential for AR nucleocytoplasmic trafficking. By characterizing the localization of green fluorescence protein (GFP)-tagged NES(AR), we designed and executed a yeast mutagenesis screen and isolated 7 yeast mutants that failed to display the NES(AR) export function. One of those mutants was identified as the splicing factor pre-mRNA processing factor 8 (Prp8). We further showed that Prp8 could regulate NES(AR) function using short hairpin RNA knockdown of Prp8 coupled with a rapamycin export assay in mammalian cells and knockdown of Prp8 could induce nuclear accumulation of GFP-tagged AR in PC3 cells. Prp8 expression was decreased in castration-resistant LuCaP35 xenograft tumors as compared with androgen-sensitive xenografts. Laser capture microdissection and quantitative PCR showed Prp8 mRNA levels were decreased in human prostate cancer specimens with high Gleason scores. In prostate cancer cells, coimmunoprecipitation and deletion mutagenesis revealed a physical interaction between Prp8 and AR mainly mediated by NES(AR). Luciferase assay with prostate specific antigen promoter-driven reporter demonstrated that Prp8 regulated AR transcription activity in prostate cancer cells. Interestingly, Prp8 knockdown also increased polyubiquitination of endogenous AR. This may be 1 possible mechanism by which it modulates AR activity. These results show that Prp8 is a novel AR cofactor that interacts with NES(AR) and regulates AR function in prostate cancer cells.


Assuntos
Sinais de Exportação Nuclear/fisiologia , Neoplasias da Próstata/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores Androgênicos/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/metabolismo , Ribonucleoproteína Nuclear Pequena U5/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Humanos , Técnicas In Vitro , Masculino , Próstata/metabolismo , Próstata/patologia , Ligação Proteica , Transdução de Sinais/fisiologia
9.
Biochem J ; 471(2): 199-209, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26268559

RESUMO

As an orphan member of the nuclear receptor family, liver receptor homologue-1 (LRH-1) controls a tremendous range of transcriptional programmes that are essential for metabolism and hormone synthesis. Our previous studies have shown that nuclear localization of the LRH-1 protein is mediated by two nuclear localization signals (NLSs) that are karyopherin/importin-dependent. It is unclear whether LRH-1 can be actively exported from the nucleus to the cytoplasm. In the present study, we describe a nuclear export domain containing two leucine-rich motifs [named nuclear export signal (NES)1 and NES2] within the ligand-binding domain (LBD). Mutation of leucine residues in NES1 or NES2 abolished nuclear export, indicating that both NES1 and NES2 motifs are essential for full nuclear export activity. This NES-mediated nuclear export was insensitive to the chromosomal region maintenance 1 (CRM1) inhibitor leptomycin B (LMB) or to CRM1 knockdown. However, knockdown of calreticulin (CRT) prevented NES-mediated nuclear export. Furthermore, our data show that CRT interacts with LRH-1 and is involved in the nuclear export of LRH-1. With full-length LRH-1, mutation of NES1 led to perinuclear accumulation of the mutant protein. Immunofluorescence analysis showed that these perinuclear aggregates were co-localized with the centrosome marker, microtubule-associated protein 1 light chain 3 (LC3), ubiquitin and heat shock protein 70 (Hsp70), indicating that the mutant was misfolded and sequestered into aggresome-like structures via the autophagic clearance pathway. Our study demonstrates for the first time that LRH-1 has a CRT-dependent NES which is not only required for cytoplasmic trafficking, but also essential for correct protein folding to avoid misfolding-induced aggregation.


Assuntos
Calbindina 2/metabolismo , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Sinais de Exportação Nuclear/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Células COS , Calbindina 2/genética , Núcleo Celular/genética , Chlorocebus aethiops , Citoplasma/genética , Proteínas de Choque Térmico HSP70/genética , Proteínas de Choque Térmico HSP70/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Receptores Citoplasmáticos e Nucleares/genética
10.
Virology ; 474: 28-33, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25463601

RESUMO

We recently determined that the nuclear import of cutaneous beta genus HPV8 E7 oncoprotein it is mediated by its zinc-binding domain via direct hydrophobic interactions with the FG nucleoporins Nup62 and Nup153 (Onder and Moroianu, 2014). Here we investigated the nuclear export of HPV8 E7 oncoprotein using confocal microscopy after transfections of HeLa cells with EGFP-8cE7 and mutant plasmids and treatment with Ratjadone A nuclear export inhibitor. We determined that HPV8 E7 contains a leucine-rich nuclear export signal (NES), 76IRTFQELLF84, within its zinc-binding domain that mediates its nuclear export via a CRM1 pathway. We found that HPV8 E7 interacts with CRM1 and that the hydrophobic amino acid residues I76, F79 and L82 of the NES are essential for this interaction and for nuclear export of HPV8 E7 oncoprotein.


Assuntos
Betapapillomavirus/metabolismo , Carioferinas/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transporte Ativo do Núcleo Celular , Substituição de Aminoácidos , Betapapillomavirus/genética , Betapapillomavirus/patogenicidade , Células HeLa , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Mutagênese Sítio-Dirigida , Sinais de Exportação Nuclear/genética , Sinais de Exportação Nuclear/fisiologia , Proteínas E7 de Papillomavirus/química , Proteínas E7 de Papillomavirus/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/virologia , Proteína Exportina 1
11.
PLoS One ; 9(12): e114629, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25485706

RESUMO

The hepatitis C virus (HCV) is a positive strand RNA virus of the Flavivirus family that replicates in the cytoplasm of infected hepatocytes. Previously, several nuclear localization signals (NLS) and nuclear export signals (NES) have been identified in HCV proteins, however, there is little evidence that these proteins travel into the nucleus during infection. We have recently shown that nuclear pore complex (NPC) proteins (termed nucleoporins or Nups) are present in the membranous web and are required during HCV infection. In this study, we identify a total of 11 NLS and NES sequences in various HCV proteins. We show direct interactions between HCV proteins and importin α5 (IPOA5/kapα1), importin ß3 (IPO5/kap ß3), and exportin 1 (XPO1/CRM1) both in-vitro and in cell culture. These interactions can be disrupted using peptides containing the specific NLS or NES sequences of HCV proteins. Moreover, using a synchronized infection system, we show that these peptides inhibit HCV infection during distinct phases of the HCV life cycle. The inhibitory effects of these peptides place them in two groups. The first group binds IPOA5 and inhibits infection during the replication stage of HCV life cycle. The second group binds IPO5 and is active during both early replication and early assembly. This work delineates the entire life cycle of HCV and the active involvement of NLS sequences during HCV replication and assembly. Given the abundance of NLS sequences within HCV proteins, our previous finding that Nups play a role in HCV infection, and the relocation of the NLS double-GFP reporter in HCV infected cells, this work supports our previous hypothesis that NPC-like structures and nuclear transport factors function in the membranous web to create an environment conducive to viral replication.


Assuntos
Membrana Celular/fisiologia , Hepacivirus/fisiologia , Antígenos da Hepatite C/metabolismo , Hepatite C/metabolismo , Sinais de Exportação Nuclear/fisiologia , Sinais de Localização Nuclear/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas do Core Viral/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Sequência de Aminoácidos , Western Blotting , Núcleo Celular/metabolismo , Proliferação de Células , Células Cultivadas , Citoplasma/metabolismo , Hepatite C/genética , Hepatite C/virologia , Antígenos da Hepatite C/genética , Humanos , Dados de Sequência Molecular , Sinais de Localização Nuclear/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Fragmentos de Peptídeos/farmacologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas do Core Viral/genética , Replicação Viral
12.
PLoS Comput Biol ; 10(9): e1003841, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25233087

RESUMO

The nuclear export of proteins is regulated largely through the exportin/CRM1 pathway, which involves the specific recognition of leucine-rich nuclear export signals (NESs) in the cargo proteins, and modulates nuclear-cytoplasmic protein shuttling by antagonizing the nuclear import activity mediated by importins and the nuclear import signal (NLS). Although the prediction of NESs can help to define proteins that undergo regulated nuclear export, current methods of predicting NESs, including computational tools and consensus-sequence-based searches, have limited accuracy, especially in terms of their specificity. We found that each residue within an NES largely contributes independently and additively to the entire nuclear export activity. We created activity-based profiles of all classes of NESs with a comprehensive mutational analysis in mammalian cells. The profiles highlight a number of specific activity-affecting residues not only at the conserved hydrophobic positions but also in the linker and flanking regions. We then developed a computational tool, NESmapper, to predict NESs by using profiles that had been further optimized by training and combining the amino acid properties of the NES-flanking regions. This tool successfully reduced the considerable number of false positives, and the overall prediction accuracy was higher than that of other methods, including NESsential and Wregex. This profile-based prediction strategy is a reliable way to identify functional protein motifs. NESmapper is available at http://sourceforge.net/projects/nesmapper.


Assuntos
Biologia Computacional/métodos , Leucina/química , Sinais de Exportação Nuclear/genética , Análise de Sequência de Proteína/métodos , Transporte Ativo do Núcleo Celular/genética , Transporte Ativo do Núcleo Celular/fisiologia , Sequência de Aminoácidos , Animais , Análise Mutacional de DNA , Leucina/genética , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Sinais de Exportação Nuclear/fisiologia , Curva ROC , Software
13.
J Virol ; 88(13): 7455-63, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24741105

RESUMO

UNLABELLED: The influenza A virus nuclear export protein (NEP) plays crucial roles in the nuclear export of the viral ribonucleoprotein complex through the chromosome region maintenance 1 (CRM1)-mediated cellular protein transport system. However, the detailed mechanism of NEP nucleocytoplasmic trafficking remains incompletely understood. Here, we investigated the subcellular localization of NEP from two strains of H1N1 influenza A virus and found that 2009 swine-origin H1N1 influenza A virus A/California/04/2009 (CA04) NEP displayed a distinct cellular distribution pattern, forming unique nuclear aggregates, compared to A/WSN/33 (H1N1) (WSN) NEP. Characterization of the nucleocytoplasmic transport pathways of these two NEPs showed that they both enter the nucleus by passive diffusion but are exported through the nuclear export receptor CRM1-mediated pathway with different efficiencies. The two identified nuclear export signals (NESs) on the two NEPs functioned similarly despite differences in their amino acid sequences. Using a two-hybrid assay, we confirmed that the CA04 NEP interacts less efficiently with CRM1 and that a threonine residue at position 48 is responsible for the nuclear aggregation. The present study revealed the dissimilarity in subcellular NEP transport processes between the 2009 pandemic (H1N1) influenza A virus CA04 and the laboratory-adapted H1N1 virus WSN and uncovered the mechanism responsible for this difference. IMPORTANCE: Because the efficiency of the nucleocytoplasmic transport of viral components is often correlated with the viral RNA polymerase activity, propagation, and host range of influenza viruses, the present study investigated the subcellular localization of NEP from two strains of H1N1 influenza virus. We found that the NEPs of both A/California/04/2009 (H1N1) (CA04) and A/WSN/33 (H1N1) (WSN) enter the nucleus by passive diffusion but are exported with different efficiencies, which were caused by weaker binding activity between the CA04 NEP and CRM1. The results of the present study revealed characteristics of the nuclear import and export pathways of NEP and the mechanism responsible for the differences in the cellular distribution of NEP between two H1N1 strains.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/metabolismo , Carioferinas/metabolismo , Sinais de Exportação Nuclear/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Núcleo Celular/metabolismo , Humanos , Vírus da Influenza A Subtipo H1N1/classificação , Influenza Humana/virologia , Microscopia de Fluorescência , Frações Subcelulares , Técnicas do Sistema de Duplo-Híbrido , Replicação Viral/fisiologia , Proteína Exportina 1
14.
J Mol Biol ; 425(2): 350-64, 2013 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-23164569

RESUMO

CRM1 mediates nuclear export of numerous proteins and ribonucleoproteins containing a leucine-rich nuclear export signal (NES). Binding of RanGTP to CRM1 in the nucleus stabilizes cargo association with CRM1, and vice versa, but the mechanism underlying the positive cooperativity in RanGTP and NES binding to CRM1 remains incompletely understood. Herein we report a 2.1-Å-resolution crystal structure of unliganded Saccharomyces cerevisiae CRM1 (Xpo1p) that demonstrates that an internal loop of CRM1 (referred to as HEAT9 loop) is primarily responsible for maintaining the NES-binding cleft in a closed conformation, rendering CRM1 incapable of NES binding in the absence of RanGTP. The structure also shows that the C-terminal tail of CRM1 stabilizes the autoinhibitory conformation of the HEAT9 loop and thereby reinforces autoinhibition. Comparison with the structures of CRM1-NES-RanGTP complexes reveals how binding of RanGTP is associated with a series of allosteric conformational changes in CRM1 that lead to opening of the NES-binding cleft, allowing for stable binding of NES cargoes.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Núcleo Celular/metabolismo , Carioferinas/química , Sinais de Exportação Nuclear/fisiologia , Receptores Citoplasmáticos e Nucleares/química , Saccharomyces cerevisiae/metabolismo , Regulação Alostérica , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Carioferinas/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Receptores Citoplasmáticos e Nucleares/metabolismo , Homologia de Sequência de Aminoácidos , Proteína ran de Ligação ao GTP/metabolismo , Proteína Exportina 1
15.
RNA ; 19(1): 17-35, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23150874

RESUMO

GW182 family proteins play important roles in microRNA (miRNA)-mediated gene silencing. They interact with Argonaute (Ago) proteins and localize in processing bodies, which are cytoplasmic foci involved in mRNA degradation and storage. Here, we demonstrated that human GW182 paralog, TNRC6A, is a nuclear-cytoplasmic shuttling protein, and its subcellular localization is conducted by a nuclear export signal (NES) and a nuclear localization signal (NLS) identified in this study. TNRC6A with mutations in its NES region was predominantly localized in the nucleus in an Ago-independent manner. However, it was found that TNRC6A could bring Ago protein into the nucleus via its Ago-interacting motif(s). Furthermore, miRNAs were also colocalized with nuclear TNRC6A-Ago and exhibited gene silencing activity. These results proposed the possibility that TNRC6A plays an important role in navigating Ago protein into the nucleus to lead miRNA-mediated gene silencing.


Assuntos
Proteínas Argonautas/metabolismo , Autoantígenos/metabolismo , Núcleo Celular/metabolismo , Inativação Gênica , MicroRNAs/metabolismo , Proteínas de Ligação a RNA/metabolismo , Motivos de Aminoácidos , Autoantígenos/genética , Núcleo Celular/genética , Células HeLa , Humanos , Mutação , Sinais de Exportação Nuclear/genética , Sinais de Exportação Nuclear/fisiologia , Sinais de Localização Nuclear/genética , Sinais de Localização Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Proteínas de Ligação a RNA/genética
16.
J Biol Chem ; 287(30): 25501-9, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22692200

RESUMO

Chk1 plays a key role in regulating the replication checkpoint and DNA damage response. Recent evidence suggests that mammalian Chk1 regulates both the nuclear and cytoplasmic checkpoint events. However, mechanisms regulating cellular mobilization of Chk1 were not well understood. Here, we report the identification of regions of human Chk1 that regulate its protein cellular localization and checkpoint function. We demonstrate that the two highly conserved motifs (CM1 and CM2) at the C terminus of Chk1 function as a nuclear export signal and nuclear localization signal, respectively. Mutating five highly conserved residues within these two motifs of Chk1 resulted in its accumulation mainly in the cytoplasm. These cytoplasmic Chk1 mutants were less stable and exhibited significantly reduced phosphorylation by DNA damage treatment, yet they retained, at least partially, checkpoint function. Using an adenovirus-mediated gene targeting technique, we attempted to create an HCT116 cell line in which endogenous Chk1 is mutated so that it is expressed exclusively in the cytoplasm. However, we failed to obtain homozygous mutant cell lines. We found that even the heterozygous mutant cell lines showed cell survival defects accompanied by spontaneous cell death. Together, these results reveal novel regulatory mechanisms that couple protein cellular localization with the checkpoint response and cell viability of Chk1.


Assuntos
Pontos de Checagem do Ciclo Celular/fisiologia , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Sinais de Exportação Nuclear/fisiologia , Proteínas Quinases/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Motivos de Aminoácidos , Núcleo Celular/genética , Sobrevivência Celular/fisiologia , Quinase 1 do Ponto de Checagem , Citoplasma/genética , Células HEK293 , Células HeLa , Heterozigoto , Humanos , Mutação , Proteínas Quinases/genética
17.
PLoS One ; 7(6): e38740, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22715410

RESUMO

BACKGROUND: Human SA/STAG proteins, homologues of the yeast Irr1/Scc3 cohesin, are the least studied constituents of the sister chromatid cohesion complex crucial for proper chromosome segregation. The two SA paralogues, SA1 and SA2, show some specificity towards the chromosome region they stabilize, and SA2, but not SA1, has been shown to participate in transcriptional regulation as well. The molecular basis of this functional divergence is unknown. METHODOLOGY/PRINCIPAL FINDINGS: In silico analysis indicates numerous putative nuclear localization (NLS) and export (NES) signals in the SA proteins, suggesting the possibility of their nucleocytoplasmic shuttling. We studied the functionality of those putative signals by expressing fluorescently tagged SA1 and SA2 in the yeast Saccharomyces cerevisiae. Only the N-terminal NLS turned out to be functional in SA1. In contrast, the SA2 protein has at least two functional NLS and also two functional NES. Depending on the balance between these opposing signals, SA2 resides in the nucleus or is distributed throughout the cell. Validation of the above conclusions in HeLa cells confirmed that the same N-terminal NLS of SA1 is functional in those cells. In contrast, in SA2 the principal NLS functioning in HeLa cells is different from that identified in yeast and is localized to the C-terminus. CONCLUSIONS/SIGNIFICANCE: This is the first demonstration of the possibility of non-nuclear localization of an SA protein. The reported difference in the organization between the two SA homologues may also be relevant to their partially divergent functions. The mechanisms determining subcellular localization of cohesins are only partially conserved between yeast and human cells.


Assuntos
Antígenos Nucleares/biossíntese , Sinais de Exportação Nuclear/fisiologia , Sinais de Localização Nuclear/biossíntese , Proteínas Nucleares/biossíntese , Saccharomyces cerevisiae/metabolismo , Antígenos Nucleares/genética , Proteínas de Ciclo Celular , Humanos , Sinais de Localização Nuclear/genética , Proteínas Nucleares/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Saccharomyces cerevisiae/genética
18.
Mol Biol Cell ; 23(14): 2755-69, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22621903

RESUMO

We identified a new calmodulin kinase I (CaMKI) substrate, cytidyltransferase (CCTα), a crucial enzyme required for maintenance of cell membranes. CCTα becomes activated with translocation from the cytoplasm to the nuclear membrane, resulting in increased membrane phospholipids. Calcium-activated CCTα nuclear import is mediated by binding of its C-terminus to 14-3-3 ζ, a regulator of nuclear trafficking. Here CaMK1 phosphorylates residues within this C-terminus that signals association of CCTα with 14-3-3 ζ to initiate calcium-induced nuclear entry. CaMKI docks within the CCTα membrane-binding domain (residues 290-299), a sequence that displays similarities to a canonical nuclear export signal (NES) that also binds CRM1/exportin 1. Expression of a CFP-CCTα mutant lacking residues 290-299 in cells results in cytosolically retained enzyme. CRM1/exportin 1 was required for CCTα nuclear export, and its overexpression in cells was partially sufficient to trigger CCTα nuclear export despite calcium stimulation. An isolated CFP-290-299 peptide remained in the nucleus in the presence of leptomycin B but was able to target to the cytoplasm with farnesol. Thus CaMKI vies with CRM1/exportin 1 for access to a NES, and assembly of a CaMKI-14-3-3 ζ-CCTα complex is a key effector mechanism that drives nuclear CCTα translocation.


Assuntos
Transporte Ativo do Núcleo Celular , Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/metabolismo , Chaperonina com TCP-1/metabolismo , Carioferinas/metabolismo , Sinais de Exportação Nuclear/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas 14-3-3/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Farneseno Álcool , Ácidos Graxos Insaturados/farmacologia , Células HeLa , Humanos , Camundongos , Membrana Nuclear/metabolismo , Sinais de Localização Nuclear/metabolismo , Fosfolipídeos/biossíntese , Transporte Proteico , Transdução de Sinais , Proteína Exportina 1
19.
PLoS One ; 7(12): e52956, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23285234

RESUMO

Ubiquitin (Ub) and ubiquitin-like (UBL) proteins regulate a diverse array of cellular pathways through covalent as well as non-covalent interactions with target proteins. Yeast protein Mdy2 (Get5) and its human homolog GdX (Ubl4a) belong to the class of UBL proteins which do not form conjugates with other proteins. Mdy2 is required for cell survival under heat stress and for efficient mating. As part of a complex with Sgt2 and Get4 it has been implicated in the biogenesis of tail-anchored proteins. Interestingly, in response to heat stress, Mdy2 protein that is predominantly localized in the nucleus co-localized with poly(A)-binding protein Pab1 to cytoplasmic stress granules suggesting that nucleocytoplasmic shuttling is of functional importance. Here we investigate the nuclear import of Mdy2, a process that is independent of the Get4/Sgt2 complex but required for stress response. Nuclear import is mediated by an N-terminal nuclear localization signal (NLS) and this process is essential for the heat stress response. In contrast, cells expressing Mdy2 lacking a nuclear export signal (NES) behave like wild type. Importantly, both Mdy2 and Mdy2-ΔNES, but not Mdy2-ΔNLS, physically interact with Pab1 and this interaction correlates with the accumulation in cytoplasmic stress granules. Thus, the nuclear history of the UBL Mdy2 appears to be essential for its function in cytoplasmic stress granules during the rapid cellular response to heat stress.


Assuntos
Resposta ao Choque Térmico/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Ubiquitina/metabolismo , Transporte Ativo do Núcleo Celular/genética , Proteínas de Transporte/genética , Grânulos Citoplasmáticos/genética , Grânulos Citoplasmáticos/metabolismo , Humanos , Proteínas de Membrana , Modelos Biológicos , Sinais de Exportação Nuclear/genética , Sinais de Exportação Nuclear/fisiologia , Sinais de Localização Nuclear/genética , Organismos Geneticamente Modificados , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/fisiologia , Deleção de Sequência , Ubiquitina/química , Ubiquitina/genética , Ubiquitina/fisiologia , Ubiquitinas/genética , Ubiquitinas/metabolismo , Ubiquitinas/fisiologia
20.
Rheum Dis Clin North Am ; 37(3): 433-52, vi-vii, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22023901

RESUMO

Denosumab (Prolia) is a fully human monoclonal antibody directed against receptor activator of nuclear factor-κB ligand (RANKL), which interferes with the formation, activation, and survival of osteoclasts. It was approved by the Food and Drug Administration in June 2010 as a new treatment for postmenopausal osteoporosis in women who are at high risk for fracture. Given its mechanism of action, it is an antiresorptive therapy that is administered as a 60-mg subcutaneous injection every 6 months. It is the first biologic antiresorptive therapy for osteoporosis, and the first osteoporosis therapy to show efficacy and safety in patients with renal impairment.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Remodelação Óssea/efeitos dos fármacos , Sinais de Exportação Nuclear/efeitos dos fármacos , Osteoporose Pós-Menopausa/tratamento farmacológico , Osteoporose Pós-Menopausa/metabolismo , Ligante RANK/metabolismo , Anticorpos Monoclonais Humanizados , Remodelação Óssea/fisiologia , Denosumab , Feminino , Humanos , Pessoa de Meia-Idade , Sinais de Exportação Nuclear/fisiologia , Ligante RANK/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...