Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 236
Filtrar
2.
Acta Pharmacol Sin ; 44(2): 367-380, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35794373

RESUMO

Disrupted redox homeostasis contributes to renal ischemia-reperfusion (IR) injury. Abundant natural products can activate nuclear factor erythroid-2-related factor 2 (Nrf2), thereby providing therapeutic benefits. Methyl eugenol (ME), an analog of the phenolic compound eugenol, has the ability to induce Nrf2 activity. In this study, we investigated the protective effects of ME against renal oxidative damage in vivo and in vitro. An IR-induced acute kidney injury (AKI) model was established in mice. ME (20 mg·kg-1·d-1, i.p.) was administered to mice on 5 consecutive days before IR surgery. We showed that ME administration significantly attenuated renal destruction, improved the survival rate, reduced excessive oxidative stress and inhibited mitochondrial lesions in AKI mice. We further demonstrated that ME administration significantly enhanced Nrf2 activity and increased the expression of downstream antioxidative molecules. Similar results were observed in vitro in hypoxia/reoxygenation (HR)-exposed proximal tubule epithelial cells following pretreatment with ME (40 µmol·L-1). In both renal oxidative damage models, ME induced Nrf2 nuclear retention in tubular cells. Using specific inhibitors (CC and DIF-3) and molecular docking, we demonstrated that ME bound to the binding pocket of AMPK with high affinity and activated the AMPK/GSK3ß axis, which in turn blocked the Nrf2 nuclear export signal. In addition, ME alleviated the development of renal fibrosis induced by nonfatal IR, which is frequently encountered in the clinic. In conclusion, we demonstrate that ME modulates the AMPK/GSK3ß axis to regulate the cytoplasmic-nuclear translocation of Nrf2, resulting in Nrf2 nuclear retention and thereby enhancing antioxidant target gene transcription that protects the kidney from oxidative damage.


Assuntos
Injúria Renal Aguda , Fator 2 Relacionado a NF-E2 , Camundongos , Animais , Fator 2 Relacionado a NF-E2/metabolismo , Eugenol/metabolismo , Eugenol/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Sinais de Exportação Nuclear , Glicogênio Sintase Quinase 3 beta/metabolismo , Simulação de Acoplamento Molecular , Estresse Oxidativo , Rim , Antioxidantes/metabolismo , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/metabolismo
3.
ACS Synth Biol ; 11(10): 3529-3533, 2022 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-36180042

RESUMO

The optogenetic tool LEXY consists of the second light oxygen voltage (LOV) domain of Avena sativa phototropin 1 mutated to contain a nuclear export signal. It allows exporting from the nucleus with blue light proteins of interest (POIs) genetically fused to it. Mutations slowing the dark recovery rate of the LOV domain within LEXY were recently shown to allow for better depletion of some POIs from the nucleus in Drosophila embryos and for the usage of low light illumination regimes. We investigated these variants in mammalian cells and found they increase the cytoplasmic localization of the proteins we tested after illumination, but also during the dark phases, which corresponds to higher leakiness of the system. These data suggest that, when aiming to sequester into the nucleus a protein with a cytoplasmic function, the original LEXY is preferable. The iLEXY variants are, instead, advantageous when wanting to deplete the nucleus of the POI as much as possible.


Assuntos
Proteínas Nucleares , Fototropinas , Animais , Fototropinas/genética , Fototropinas/metabolismo , Proteínas Nucleares/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Sinais de Exportação Nuclear/genética , Luz , Avena/genética , Avena/metabolismo , Oxigênio/metabolismo , Mamíferos/metabolismo
4.
Methods Mol Biol ; 2502: 245-256, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35412243

RESUMO

CRM1 recognizes hundreds to thousands of protein cargoes by binding to the eight to fifteen residue-long nuclear export signals (NESs) within their polypeptide chains. Various assays to measure the binding affinity of NESs for CRM1 have been developed. CRM1 binds to NESs with a wide range of binding affinities, with dissociation constants that span from low nanomolar to tens of micromolar. An optimized binding affinity assay with improved throughput was recently developed to measure binding affinities of NES peptides for CRM1 in the presence of excess RanGTP. The assay can measure affinities, with multiple replicates, for up to seven different NES peptides per screening plate. Here, we present a protocol for the purification of the necessary proteins and for measuring CRM1-NES binding affinities.


Assuntos
Carioferinas , Sinais de Exportação Nuclear , Receptores Citoplasmáticos e Nucleares , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , Carioferinas/química , Carioferinas/metabolismo , Peptídeos/metabolismo , Ligação Proteica , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteína Exportina 1
5.
Traffic ; 22(12): 482-489, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34622522

RESUMO

Although the majority of viruses of the family Mononegvirales replicate exclusively in the host cell cytoplasm, many of these viruses encode proteins that traffic between the nucleus and cytoplasm, which is believed to enable accessory functions in modulating the biology of the infected host cell. Among these, the P3 protein of rabies virus localizes to the nucleus through the activity of several specific nuclear localization and nuclear export signals. The major defined functions of P3 are in evasion of interferon (IFN)-mediated antiviral responses, including through inhibition of DNA-binding by IFN-activated STAT1. P3 also localizes to nucleoli and promyelocytic leukemia (PML) nuclear bodies, and interacts with nucleolin and PML protein, indicative of several intranuclear roles. The relationship of P3 nuclear localization with pathogenicity, however, is unresolved. We report that nucleocytoplasmic localization of P3 proteins from a pathogenic RABV strain, Nishigahara (Ni) and a non-pathogenic Ni-derived strain, Ni-CE, differs significantly, with nuclear accumulation defective for Ni-CE-P3. Molecular mapping indicates that altered localization derives from a coordinated effect, including two residue substitutions that independently disable nuclear localization and augment nuclear export signals, collectively promoting nuclear exclusion. Intriguingly, this appears to relate to effects on protein conformation or regulatory mechanisms, rather than direct modification of defined trafficking signal sequences. These data provide new insights into the role of regulated nuclear trafficking of a viral protein in the pathogenicity of a virus that replicates in the cytoplasm.


Assuntos
Vírus da Raiva , Núcleo Celular/metabolismo , Sinais de Exportação Nuclear , Vírus da Raiva/metabolismo , Proteínas Virais/metabolismo , Virulência
6.
Genes (Basel) ; 12(9)2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34573408

RESUMO

Nucleophosmin (NPM1) mutations occurring in acute myeloid leukemia (AML) (about 50 so far identified) cluster almost exclusively in exon 12 and lead to common changes at the NPM1 mutants C-terminus, i.e., loss of tryptophans 288 and 290 (or 290 alone) and creation of a new nuclear export signal (NES), at the bases of exportin-1(XPO1)-mediated aberrant cytoplasmic NPM1. Immunohistochemistry (IHC) detects cytoplasmic NPM1 and is predictive of the molecular alteration. Besides IHC and molecular sequencing, Western blotting (WB) with anti-NPM1 mutant specific antibodies is another approach to identify NPM1-mutated AML. Here, we show that among 382 AML cases with NPM1 exon 12 mutations, one was not recognized by WB, and describe the discovery of a novel combination of two mutations involving exon 12. This appeared as a conventional mutation A with the known TCTG nucleotides insertion/duplication accompanied by a second event (i.e., an 8-nucleotide deletion occurring 15 nucleotides downstream of the TCTG insertion), resulting in a new C-terminal protein sequence. Strikingly, the sequence included a functional NES ensuring cytoplasmic relocation of the new mutant supporting the role of cytoplasmic NPM1 as critical in AML leukemogenesis.


Assuntos
Leucemia Mieloide Aguda , Sinais de Exportação Nuclear/genética , Nucleofosmina/genética , Transporte Ativo do Núcleo Celular/genética , Idoso , Animais , Células Cultivadas , Citoplasma/metabolismo , Humanos , Imuno-Histoquímica , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Mutação , Células NIH 3T3 , Nucleofosmina/química , Nucleofosmina/metabolismo , Transporte Proteico/genética
7.
Int Immunopharmacol ; 99: 107624, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34343939

RESUMO

Hepatocellular carcinoma (HCC) is an inflammation-induced and chemotherapy-resistant common liver cancer, and a major cause of death. Some natural products have been found to be used as photosensitizers in photodynamic therapy of HCC. Due to its specific molecular structure diversities and biological activities, current status of HCC treatment with nature production remains unsatisfactory, owing largely to the toxicity, side effect and inefficiency to drug targeting. Herein, we show a nanoparticle-based broad-spectrum anti-inflammatory strategy that naïve neutrophil membrane-coated PLGA nanoparticles (NM-HB NPs) were constructed for synchronous nearinfrared fluorescence (NIR FL) imaging and photodynamic therapy (PDT) for HCC. Moreover, NM-HB NPs inhibited the expression of JUNB and promoted the ROS production. JUNB depletion enhanced the anti-HCC effect of NM-HB NPs. Importantly, it was shown that NM-HB NPs are well targeted to the tumor site and overcomes the blood circulation and immune elimination in vivo and vitro. In a mouse model of HCC, the neutrophil membrane-coated nanoparticles (NM-HB NPs) show significant therapeutic efficacy by PDT and suppressing tumor tissue increase. All results demonstrated that NM coated HB NPs representing a viable and effective treatment option for HCC.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Membrana Celular , Neoplasias Hepáticas/tratamento farmacológico , Nanopartículas/administração & dosagem , Neutrófilos , Perileno/análogos & derivados , Quinonas/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/sangue , Carcinoma Hepatocelular/patologia , Linhagem Celular , Humanos , Interleucina-6/sangue , Interleucina-6/genética , Neoplasias Hepáticas/sangue , Neoplasias Hepáticas/patologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Sinais de Exportação Nuclear , Perileno/administração & dosagem , Fotoquimioterapia , Ratos , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Carga Tumoral/efeitos dos fármacos , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/genética
8.
Int J Mol Sci ; 22(5)2021 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-33808029

RESUMO

Members of the human Zyxin family are LIM domain-containing proteins that perform critical cellular functions and are indispensable for cellular integrity. Despite their importance, not much is known about their structure, functions, interactions and dynamics. To provide insights into these, we used a set of in-silico tools and databases and analyzed their amino acid sequence, phylogeny, post-translational modifications, structure-dynamics, molecular interactions, and functions. Our analysis revealed that zyxin members are ohnologs. Presence of a conserved nuclear export signal composed of LxxLxL/LxxxLxL consensus sequence, as well as a possible nuclear localization signal, suggesting that Zyxin family members may have nuclear and cytoplasmic roles. The molecular modeling and structural analysis indicated that Zyxin family LIM domains share similarities with transcriptional regulators and have positively charged electrostatic patches, which may indicate that they have previously unanticipated nucleic acid binding properties. Intrinsic dynamics analysis of Lim domains suggest that only Lim1 has similar internal dynamics properties, unlike Lim2/3. Furthermore, we analyzed protein expression and mutational frequency in various malignancies, as well as mapped protein-protein interaction networks they are involved in. Overall, our comprehensive bioinformatic analysis suggests that these proteins may play important roles in mediating protein-protein and protein-nucleic acid interactions.


Assuntos
Biologia Computacional , Sinais de Exportação Nuclear , Zixina , Humanos , Domínios Proteicos , Transporte Proteico , Relação Estrutura-Atividade , Zixina/química , Zixina/genética , Zixina/metabolismo
9.
Int J Nanomedicine ; 16: 2833-2847, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33883894

RESUMO

INTRODUCTION: Peptides can be rationally designed as non-covalent inhibitors for molecularly targeted therapy. However, it remains challenging to efficiently deliver the peptides into the targeted cells, which often severely affects their therapeutic efficiency. METHODS: Herein, we created a novel non-covalent peptide inhibitor against nuclear export factor CRM1 by a structure-guided drug design method and targetedly delivered the peptide into cancer cells by a nanoparticle-mediated gene expression system for use as a cancer therapy. RESULTS: The nuclear export signal (NES)-optimized CRM1 peptide inhibitor colocalized with CRM1 to the nuclear envelope and inhibited nuclear export in cancer cell lines in vitro. The crystal structures of the inhibitors complexed with CRM1 were solved. In contrast to the covalent inhibitors, the peptides were similarly effective against cells harboring the CRM1 C528S mutation. Moreover, a plasmid encoding the peptides was delivered by a iRGD-modified nanoparticle to efficiently target and transfect the cancer cells in vivo after intravenous administration. The peptides could be selectively expressed in the tumor, resulting in the efficient inhibition of subcutaneous melanoma xenografts without obvious systemic toxicity. DISCUSSION: This work provides an effective strategy to design peptide-based molecularly targeted therapeutics, which could lead to the development of future targeted therapy.


Assuntos
Espaço Intracelular/metabolismo , Carioferinas/antagonistas & inibidores , Melanoma Experimental/tratamento farmacológico , Nanopartículas/química , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Células A549 , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Sequência de Aminoácidos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Carioferinas/química , Carioferinas/metabolismo , Melanoma Experimental/patologia , Proteínas Mutantes/metabolismo , Mutação/genética , Nanopartículas/ultraestrutura , Sinais de Exportação Nuclear , Peptídeos/química , Ligação Proteica/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas não Estruturais Virais/química , Proteína Exportina 1
10.
Nat Commun ; 12(1): 1505, 2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33686072

RESUMO

Survivin's dual function as apoptosis inhibitor and regulator of cell proliferation is mediated via its interaction with the export receptor CRM1. This protein-protein interaction represents an attractive target in cancer research and therapy. Here, we report a sophisticated strategy addressing Survivin's nuclear export signal (NES), the binding site of CRM1, with advanced supramolecular tweezers for lysine and arginine. These were covalently connected to small peptides resembling the natural, self-complementary dimer interface which largely overlaps with the NES. Several biochemical methods demonstrated sequence-selective NES recognition and interference with the critical receptor interaction. These data were strongly supported by molecular dynamics simulations and multiscale computational studies. Rational design of lysine tweezers equipped with a peptidic recognition element thus allowed to address a previously unapproachable protein surface area. As an experimental proof-of-principle for specific transport signal interference, this concept should be transferable to any protein epitope with a flanking well-accessible lysine.


Assuntos
Carioferinas/química , Carioferinas/metabolismo , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Survivina/química , Survivina/metabolismo , Sítios de Ligação , Proliferação de Células , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Modelos Moleculares , Sinais de Exportação Nuclear , Ligação Proteica , Conformação Proteica , Proteína Exportina 1
11.
Acta Crystallogr F Struct Biol Commun ; 77(Pt 3): 70-78, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33682791

RESUMO

CRM1 is a nuclear export receptor that has been intensively targeted over the last decade for the development of antitumor and antiviral drugs. Structural analysis of several inhibitor compounds bound to CRM1 revealed that their mechanism of action relies on the covalent modification of a critical cysteine residue (Cys528 in the human receptor) located in the nuclear export signal-binding cleft. This study presents the crystal structure of human CRM1, covalently modified by 2-mercaptoethanol on Cys528, in complex with RanGTP at 2.58 Šresolution. The results demonstrate that buffer components can interfere with the characterization of cysteine-dependent inhibitor compounds.


Assuntos
Cisteína/química , Carioferinas/química , Carioferinas/metabolismo , Mercaptoetanol/química , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteína ran de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Humanos , Modelos Moleculares , Sinais de Exportação Nuclear , Proteína ran de Ligação ao GTP/química , Proteína Exportina 1
12.
Oncogene ; 40(12): 2243-2257, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33649538

RESUMO

Mdm2 antagonizes the tumor suppressor p53. Targeting the Mdm2-p53 interaction represents an attractive approach for the treatment of cancers with functional p53. Investigating mechanisms underlying Mdm2-p53 regulation is therefore important. The scaffold protein ß-arrestin2 (ß-arr2) regulates tumor suppressor p53 by counteracting Mdm2. ß-arr2 nucleocytoplasmic shuttling displaces Mdm2 from the nucleus to the cytoplasm resulting in enhanced p53 signaling. ß-arr2 is constitutively exported from the nucleus, via a nuclear export signal, but mechanisms regulating its nuclear entry are not completely elucidated. ß-arr2 can be SUMOylated, but no information is available on how SUMO may regulate ß-arr2 nucleocytoplasmic shuttling. While we found ß-arr2 SUMOylation to be dispensable for nuclear import, we identified a non-covalent interaction between SUMO and ß-arr2, via a SUMO interaction motif (SIM), that is required for ß-arr2 cytonuclear trafficking. This SIM promotes association of ß-arr2 with the multimolecular RanBP2/RanGAP1-SUMO nucleocytoplasmic transport hub that resides on the cytoplasmic filaments of the nuclear pore complex. Depletion of RanBP2/RanGAP1-SUMO levels result in defective ß-arr2 nuclear entry. Mutation of the SIM inhibits ß-arr2 nuclear import, its ability to delocalize Mdm2 from the nucleus to the cytoplasm and enhanced p53 signaling in lung and breast tumor cell lines. Thus, a ß-arr2 SIM nuclear entry checkpoint, coupled with active ß-arr2 nuclear export, regulates its cytonuclear trafficking function to control the Mdm2-p53 signaling axis.


Assuntos
Proteínas Ativadoras de GTPase/genética , Neoplasias/genética , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteína SUMO-1/genética , Proteína Supressora de Tumor p53/genética , beta-Arrestina 2/genética , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Humanos , Mutação/genética , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Sinais de Exportação Nuclear/genética , Transdução de Sinais/genética , Sumoilação/genética
13.
Cell Rep ; 34(1): 108586, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33406424

RESUMO

The cyclic GMP-AMP (cGAMP) synthase (cGAS) is a key DNA sensor that initiates STING-dependent signaling to produce type I interferons through synthesizing the secondary messenger 2'3'-cGAMP. In this study, we confirm previous studies showing that cGAS is located both in the cytoplasm and in the nucleus. Nuclear accumulation is observed when leptomycin B is used to block the exportin, CRM1 protein. As a result, leptomycin B impairs the production of interferons in response to DNA stimulation. We further identify a functional nuclear export signal (NES) in cGAS, 169LEKLKL174. Mutating this NES leads to the sequestration of cGAS within the nucleus and the loss of interferon response to cytosolic DNA treatment, and it further determines the key amino acid to L172. Collectively, our data demonstrate that the cytosolic DNA-sensing function of cGAS depends on its presence within the cytoplasm, which is warranted by a functional NES.


Assuntos
Citosol/metabolismo , DNA/metabolismo , Carioferinas/metabolismo , Sinais de Exportação Nuclear , Nucleotídeos Cíclicos/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Sequência de Aminoácidos , Ácidos Graxos Insaturados/farmacologia , Células HEK293 , Células HeLa , Humanos , Imunidade Inata , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/metabolismo , Interferon beta/metabolismo , Carioferinas/efeitos dos fármacos , Carioferinas/imunologia , Proteínas de Membrana/metabolismo , Mutação , Nucleotidiltransferases/imunologia , Receptores Citoplasmáticos e Nucleares/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/imunologia , Transdução de Sinais , Células THP-1 , Proteína Exportina 1
14.
Biochim Biophys Acta Mol Cell Res ; 1868(1): 118862, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32979422

RESUMO

Spastin, a microtubule-severing AAA ATPase, regulates microtubule dynamics and plays important roles in cell division and neurogenesis. Mutations in the spastin-coding gene SPAST lead to neurodegenerative disorders and cause spastic paraplegia type 4. Spastin has two main isoforms, M1 and M87, that differ only in the presence or absence of 86 N-terminal amino acids and have alternative splicing variants that lack exon4. The N-terminal region of M1 contains a hydrophobic domain, nuclear localization signal (NLS), and nuclear export signal (NES), which partly explains the differences in the two isoforms' localization. However, the mechanisms involved in regulating isoform localization, and the effects of localization on spastin functions are not fully understood. We found endogenous M1 and M87 shuttled between the nucleus and cytoplasm during the cell cycle. We identified a NES (amino acids 195-204) that spans the microtubule-interacting and endosomal-trafficking domain and exon4 region. Furthermore, the NES sequence contains both the coiled-coil and exon4 region of spastin isoforms. Highly conserved leucine 195 in exon3 and the two residues in exon4 are crucial for predicted coiled-coil formation. Mutations in NES or leptomycin B treatment reduced cytoplasmic localization and microtubule fragmentation in M87 rather than in M1. Phosphomimetic mutation of threonine 306 adjacent to the NLS (amino acids 309-312) inhibited nuclear transport of M87. Our results indicate that the newly identified NES in the spastin isoforms containing exon4 regulates the subcellular localization of spastin in coordination with NLS controlled by the phosphorylation state of spastin, and is involved in microtubule severing.


Assuntos
Transporte Ativo do Núcleo Celular/genética , Microtúbulos/genética , Paraplegia Espástica Hereditária/genética , Espastina/genética , Adenosina Trifosfatases/genética , Ciclo Celular/genética , Núcleo Celular/genética , Células Cultivadas , Citoplasma/genética , Éxons/genética , Humanos , Mutação/genética , Sinais de Exportação Nuclear/genética , Paraplegia Espástica Hereditária/metabolismo , Paraplegia Espástica Hereditária/patologia
15.
Int J Mol Sci ; 21(17)2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32882917

RESUMO

The nuclear export receptor CRM1 (XPO1) recognizes and binds specific sequence motifs termed nuclear export signals (NESs) in cargo proteins. About 200 NES motifs have been identified, but over a thousand human proteins are potential CRM1 cargos, and most of their NESs remain to be identified. On the other hand, the interaction of NES peptides with the "NES-binding groove" of CRM1 was studied in detail using structural and biochemical analyses, but a better understanding of CRM1 function requires further investigation of how the results from these in vitro studies translate into actual NES export in a cellular context. Here we show that a simple cellular assay, based on a recently described reporter (SRVB/A), can be applied to identify novel potential NESs motifs, and to obtain relevant information on different aspects of CRM1-mediated NES export. Using cellular assays, we first map 19 new sequence motifs with nuclear export activity in 14 cancer-related proteins that are potential CRM1 cargos. Next, we investigate the effect of mutations in individual NES-binding groove residues, providing further insight into CRM1-mediated NES export. Finally, we extend the search for CRM1-dependent NESs to a recently uncovered, but potentially vast, set of small proteins called micropeptides. By doing so, we report the first NES-harboring human micropeptides.


Assuntos
Genes Reporter , Carioferinas/metabolismo , Mutação , Proteínas de Neoplasias/metabolismo , Sinais de Exportação Nuclear , Fragmentos de Peptídeos/análise , Receptores Citoplasmáticos e Nucleares/metabolismo , Transporte Ativo do Núcleo Celular , Motivos de Aminoácidos , Células HeLa , Humanos , Carioferinas/genética , Proteínas de Neoplasias/genética , Neoplasias , Receptores Citoplasmáticos e Nucleares/genética , Proteína Exportina 1
16.
Viruses ; 12(8)2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32824614

RESUMO

Caprine arthritis-encephalitis virus (CAEV), a lentivirus, relies on the action of the Rev protein for its replication. The CAEV Rev fulfills its function by allowing the nuclear exportation of partially spliced or unspliced viral mRNAs. In this study, we characterized the nuclear and nucleolar localization signals (NLS and NoLS, respectively) and the nuclear export signal (NES) of the CAEV Rev protein. These signals are key actors in the nucleocytoplasmic shuttling of a lentiviral Rev protein. Several deletion and alanine substitution mutants were generated from a plasmid encoding the CAEV Rev wild-type protein that was fused to the enhanced green fluorescent protein (EGFP). Following cell transfection, images were captured by confocal microscopy and the fluorescence was quantified in the different cell compartments. The results showed that the NLS region is localized between amino acids (aa) 59 to 75, has a monopartite-like structure and is exclusively composed of arginine residues. The NoLS was found to be partially associated with the NLS. Finally, the CAEV Rev protein's NES mapped between aa 89 to 101, with an aa spacing between the hydrophobic residues that was found to be unconventional as compared to that of other retroviral Rev/Rev-like proteins.


Assuntos
Vírus da Artrite-Encefalite Caprina/genética , Núcleo Celular/metabolismo , Produtos do Gene rev/genética , Sinais Direcionadores de Proteínas , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Vírus da Artrite-Encefalite Caprina/metabolismo , Bovinos , Núcleo Celular/virologia , Produtos do Gene rev/metabolismo , Proteínas de Fluorescência Verde , Células HeLa , Humanos , Macrófagos/virologia , Sinais de Exportação Nuclear , Sinais de Localização Nuclear/metabolismo
17.
Biochem Biophys Res Commun ; 531(3): 335-340, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-32800339

RESUMO

The nuclear export signal (NES) endows a protein nuclear export ability. Surprisingly, our previous study shows that just the NES peptide of Schizosaccharomyces pombe Oxs1 (SpOxs1NES) can confer diamide tolerance by competing with transcription factor Pap1 for nuclear transport. This finding intrigued us to test the function of NESs from heterologous organisms. The Arabidopsis thaliana zinc finger transcription factor OXIDATIVE STRESS 2 (AtOXS2) is a nucleocytoplasmic shuttling protein and nearly all OXS2 members from maize and rice contain an NES. In this study, we find that the plant OXS2 members and their C-terminus (AT3 peptide) can confer diamide tolerance due to their NESs, and amino acids in non-conserved as well as conserved positions are necessary for the diamide tolerance. As in SpOxs1NES, the enhanced tolerance to diamide in fission yeast depends on Pap1. Like SpOxs1NES, OXS2 family NESs appear to compete for nuclear transport of the Pap1-like Arabidopsis protein bZIP10, as when overproduced in Arabidopsis protoplasts, bZIP10 is retained in the nucleus.


Assuntos
Diamida/metabolismo , Sinais de Exportação Nuclear , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Schizosaccharomyces/metabolismo , Adaptação Fisiológica , Sequência de Aminoácidos , Substituição de Aminoácidos , Núcleo Celular/metabolismo , Sequência Conservada , Peptídeos/metabolismo , Frações Subcelulares/metabolismo
18.
Proc Natl Acad Sci U S A ; 117(28): 16557-16566, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32601201

RESUMO

Influenza viruses (IV) exploit a variety of signaling pathways. Previous studies showed that the rapidly accelerated fibrosarcoma/mitogen-activated protein kinase/extracellular signal-regulated kinase (Raf/MEK/ERK) pathway is functionally linked to nuclear export of viral ribonucleoprotein (vRNP) complexes, suggesting that vRNP export is a signaling-induced event. However, the underlying mechanism remained completely enigmatic. Here we have dissected the unknown molecular steps of signaling-driven vRNP export. We identified kinases RSK1/2 as downstream targets of virus-activated ERK signaling. While RSK2 displays an antiviral role, we demonstrate a virus-supportive function of RSK1, migrating to the nucleus to phosphorylate nucleoprotein (NP), the major constituent of vRNPs. This drives association with viral matrix protein 1 (M1) at the chromatin, important for vRNP export. Inhibition or knockdown of MEK, ERK or RSK1 caused impaired vRNP export and reduced progeny virus titers. This work not only expedites the development of anti-influenza strategies, but in addition demonstrates converse actions of different RSK isoforms.


Assuntos
Vírus da Influenza A/metabolismo , Influenza Humana/virologia , Ribonucleoproteínas/metabolismo , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , Núcleo Celular/virologia , Humanos , Vírus da Influenza A/genética , Influenza Humana/genética , Influenza Humana/metabolismo , Sistema de Sinalização das MAP Quinases , Sinais de Exportação Nuclear , Ribonucleoproteínas/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo
19.
Mol Biol Cell ; 31(17): 1879-1891, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32520643

RESUMO

The E571K mutation of CRM1 is highly prevalent in some cancers, but its mechanism of tumorigenesis is unclear. Glu571 of CRM1 is located in its nuclear export signal (NES)-binding groove, suggesting that binding of select NESs may be altered. We generated HEK 293 cells with either monoallelic CRM1WT/E571K or biallelic CRM1E571K/E571K using CRISPR/Cas9. We also combined analysis of binding affinities and structures of 27 diverse NESs for wild-type and E571K CRM1 with structure-based bioinformatics. While most NESs bind the two CRM1 similarly, NESs from Mek1, eIF4E-transporter, and RPS2 showed >10-fold affinity differences. These NESs have multiple charged side chains binding close to CRM1 position 571, but this feature alone was not sufficient to predict different binding to CRM1(E571K). Consistent with eIF4E-transporter NES binding weaker to CRM1(E571K), eIF4E-transporter was mislocalized in tumor cells carrying CRM1(E571K). This serves as proof of concept that understanding how CRM1(E571K) affects NES binding provides a platform for identifying cargoes that are mislocalized in cancer upon CRM1 mutation. Finally, we showed that large affinity changes seen with some NES peptides (of Mek1 and RPS2) do not always translate to the full-length cargoes, suggesting limitations with current NES prediction methods. Therefore, comprehensive studies like ours are imperative to identify CRM1 cargoes with real pathogenic potential.


Assuntos
Carioferinas/genética , Sinais de Exportação Nuclear/genética , Receptores Citoplasmáticos e Nucleares/genética , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos/genética , Núcleo Celular/metabolismo , Cristalografia por Raios X/métodos , Células HEK293 , Humanos , Carioferinas/metabolismo , MAP Quinase Quinase 1/metabolismo , Modelos Moleculares , Mutação/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Ligação Proteica/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Ribossômicas/metabolismo , Proteína ran de Ligação ao GTP/genética , Proteína ran de Ligação ao GTP/metabolismo , Proteína Exportina 1
20.
Sci Rep ; 10(1): 9532, 2020 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-32533018

RESUMO

The newly assigned subfamily Avulavirinae in the family Paramyxoviridae includes avian paramyxoviruses (APMVs) isolated from a wide variety of avian species across the globe. Till date, 21 species of APMVs are reported and their complete genome sequences are available in GenBank. The APMV genome comprises of a single stranded, negative sense, non-segmented RNA comprising six transcriptional units (except APMV-6 with seven units) each coding for a structural protein. Additionally, by co-transcriptional RNA editing of phosphoprotein (P) gene, two mRNAs coding for accessory viral proteins, V and W, are generated along with unedited P mRNA. However, in APMV-11, the unedited mRNA codes for V protein while +2 edited mRNA translates to P protein, similar to members of subfamily Rubulavirinae in the same family. Such RNA editing in paramyxoviruses enables maximizing the coding capacity of their smaller genome. The three proteins of P gene: P, V and W, share identical N terminal but varied C terminal sequences that contribute to their unique functions. Here, we analyzed the P gene editing site, V and W sequences of all 21 APMV species known so far (55 viruses) by using bioinformatics and report their genetic variations and molecular evolution. The variations observed in the sequence and hexamer phase positions of the P gene editing sites is likely to influence the levels and relative proportions of P, V and W proteins' expressions which could explain the differences in the pathogenicity of APMVs. The V protein sequences of APMVs had conserved motifs similar to V proteins of other paramyxoviruses including the seven cysteine residues involved in MDA5 interference, STAT1 degradation and interferon antagonism. Conversely, W protein sequences of APMVs were distinct. High sequence homology was observed in both V and W proteins between strains of the same species than between species except in APMV-3 which was the most divergent APMV species. The estimates of synonymous and non-synonymous substitution rates suggested negative selection pressure on the V and W proteins within species indicating their low evolution rate. The molecular clock analysis revealed higher conservation of V protein sequence compared to W protein indicating the important role played by V protein in viral replication, pathogenesis and immune evasion. However, we speculate the genetic diversity of W proteins could impact the degree of pathogenesis, variable interferon antagonistic activity and the wide host range exhibited by APMV species. Phylogenetically, V proteins of APMVs clustered into three groups similar to the recent classification of APMVs into three new genera while no such pattern could be deciphered in the analysis of W proteins except that strains of same species grouped together. This is the first comprehensive study describing in detail the genetic variations and the molecular evolution of P gene edited, accessory viral proteins of Avian paramyxoviruses.


Assuntos
Avulavirus/genética , Evolução Molecular , Variação Genética , Edição de RNA , Proteínas Virais/genética , Animais , Sequência Conservada , Genoma Viral/genética , Sinais de Exportação Nuclear , Sinais de Localização Nuclear , Filogenia , Proteínas Virais/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA