Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Virol ; 96(17): e0111822, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-35950857

RESUMO

Being nonpathogenic to humans, rodent parvoviruses (PVs) are naturally oncolytic viruses with great potential as anti-cancer agents. As these viruses replicate in the host cell nucleus, they must gain access to the nucleus during infection. The PV minute virus of mice (MVM) and several other PVs transiently disrupt the nuclear envelope (NE) and enter the nucleus through the resulting breaks. However, the molecular basis of this unique nuclear entry pathway remains uncharacterized. In this study, we used MVM as a model to investigate the molecular mechanism by which PVs induce NE disruption during viral nuclear entry. By combining bioinformatics analyses, metabolic labeling assays, mutagenesis, and pharmacological inhibition, we identified a functional myristoylation site at the sequence 78GGKVGH83 of the unique portion of the capsid protein VP1 (VP1u) of MVM. Performing proteolytic cleavage studies with a peptide containing this myristoylation site or with purified virions, we found tryptophan at position 77 of MVM VP1u is susceptible to chymotrypsin cleavage, implying this cleavage exposes G (glycine) 78 at the N-terminus of VP1u for myristoylation. Subsequent experiments using inhibitors of myristoylation and cellular proteases with MVM-infected cells, or an imaging-based quantitative NE permeabilization assay, further indicate protein myristoylation and a chymotrypsin-like activity are essential for MVM to locally disrupt the NE during viral nuclear entry. We thus propose a model for the nuclear entry of MVM in which NE disruption is mediated by VP1u myristoylation after the intact capsid undergoes proteolytic processing to expose the required N-terminal G for myristoylation. IMPORTANCE Rodent parvoviruses (PVs), including minute virus of mice (MVM), have the ability to infect and kill cancer cells and thereby possess great potential in anti-cancer therapy. In fact, some of these viruses are currently being investigated in both preclinical studies and clinical trials to treat a wide variety of cancers. However, the detailed mechanism of how PVs enter the cell nucleus remains unknown. In this study, we for the first time demonstrated a chemical modification called "myristoylation" of a MVM protein plays an essential role in the nuclear entry of the virus. We also showed, in addition to protein myristoylation, a chymotrypsin-like activity, which may come from cellular proteasomes, is required for MVM to get myristoylated and enter the nucleus. These findings deepen our understanding on how MVM and other related PVs infect host cells and provide new insights for the development of PV-based anti-cancer therapies.


Assuntos
Proteínas do Capsídeo , Núcleo Celular , Vírus Miúdo do Camundongo , Infecções por Parvoviridae , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Núcleo Celular/virologia , Quimotripsina/metabolismo , Camundongos , Vírus Miúdo do Camundongo/fisiologia , Infecções por Parvoviridae/metabolismo , Processamento de Proteína Pós-Traducional
2.
J Hepatol ; 68(3): 441-448, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29113909

RESUMO

BACKGROUND & AIMS: Hepatitis B virus (HBV) has a DNA genome but replicates within the nucleus by reverse transcription of an RNA pregenome, which is converted to DNA in cytoplasmic capsids. Capsids in this compartment are correlated with inflammation and epitopes of the capsid protein core (Cp) are a major target for T cell-mediated immune responses. We investigated the mechanism of cytoplasmic capsid transport, which is important for infection but also for cytosolic capsid removal. METHODS: We used virion-derived capsids containing mature rcDNA (matC) and empty capsids (empC). RNA-containing capsids (rnaC) were used as a control. The investigations comprised pull-down assays for identification of cellular interaction partners, immune fluorescence microscopy for their colocalization and electron microscopy after microinjection to determine their biological significance. RESULTS: matC and empC underwent active transport through the cytoplasm towards the nucleus, while rnaC was poorly transported. We identified the dynein light chain LL1 as a functional interaction partner linking capsids to the dynein motor complex and showed that there is no compensatory transport pathway. Using capsid and dynein LL1 mutants we characterized the required domains on the capsid and LL1. CONCLUSIONS: This is the first investigation on the detailed molecular mechanism of how matC pass the cytoplasm upon infection and how empC can be actively removed from the cytoplasm into the nucleus. Considering that hepatocytes with cytoplasmic capsids are better recognized by the T cells, we hypothesize that targeting capsid DynLL1-interaction will not only block HBV infection but also stimulate elimination of infected cells. LAY SUMMARY: In this study, we identified the molecular details of HBV translocation through the cytoplasm. Our evidence offers a new drug target which could not only inhibit infection but also stimulate immune clearance of HBV infected cells.


Assuntos
Proteínas do Capsídeo/metabolismo , DNA Viral , Vírus da Hepatite B , Hepatite B , Replicação Viral/fisiologia , Transporte Biológico/imunologia , Hepatite B/imunologia , Hepatite B/virologia , Vírus da Hepatite B/genética , Vírus da Hepatite B/fisiologia , Humanos , Imunidade Celular/imunologia , Microscopia Eletrônica/métodos , Microscopia de Fluorescência/métodos , Chaperonas Moleculares , Ligação Proteica , Vírion/imunologia
3.
J Cell Sci ; 129(15): 2905-11, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27284005

RESUMO

The transport of macromolecules into the nucleus is mediated by soluble cellular receptors of the importin ß superfamily and requires the Ran-GTPase cycle. Several studies have provided evidence that there are exceptions to this canonical nuclear import pathway. Here, we report a new unconventional nuclear import mechanism exploited by the baculovirus Autographa californica multiple nucleopolyhedrovirus (AcMNPV). We found that AcMNPV nucleocapsids entered the nucleus of digitonin-permeabilized cells in the absence of exogenous cytosol or under conditions that blocked the Ran-GTPase cycle. AcMNPV contains a protein that activates the Arp2/3 complex and induces actin polymerization at one end of the rod-shaped nucleocapsid. We show that inhibitors of Arp2/3 blocked nuclear import of nucleocapsids in semi-permeabilized cells. Nuclear import of nucleocapsids was also reconstituted in purified nuclei supplemented with G-actin and Arp2/3 under actin polymerization conditions. Thus, we propose that actin polymerization drives not only migration of baculovirus through the cytoplasm but also pushes the nucleocapsid through the nuclear pore complex to enter the cell nucleus. Our findings point to a very distinct role of actin-based motility during the baculovirus infection cycle.


Assuntos
Actinas/metabolismo , Baculoviridae/metabolismo , Núcleo Celular/metabolismo , Nucleocapsídeo/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Baculoviridae/efeitos dos fármacos , Permeabilidade da Membrana Celular/efeitos dos fármacos , Citosol/efeitos dos fármacos , Citosol/metabolismo , Digitonina/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Células HeLa , Humanos , Poro Nuclear/metabolismo , Nucleocapsídeo/efeitos dos fármacos , Nucleopoliedrovírus/efeitos dos fármacos , Nucleopoliedrovírus/metabolismo , Polimerização/efeitos dos fármacos , Quinazolinas/farmacologia , Proteína ran de Ligação ao GTP/metabolismo
4.
Biochem J ; 474(24): 4091-4104, 2017 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-29054975

RESUMO

The Neurospora crassa NIT-2 transcription factor belongs to the GATA transcription factor family and plays a fundamental role in the regulation of nitrogen metabolism. Because NIT-2 acts by accessing DNA inside the nucleus, understanding the nuclear import process of NIT-2 is necessary to characterize its function. Thus, in the present study, NIT-2 nuclear transport was investigated using a combination of biochemical, cellular, and biophysical methods. A complemented strain that produced an sfGFP-NIT-2 fusion protein was constructed, and nuclear localization assessments were made under conditions that favored protein translocation to the nucleus. Nuclear translocation was also investigated using HeLa cells, which showed that the putative NIT-2 nuclear localization sequence (NLS; 915TISSKRQRRHSKS927) was recognized by importin-α and that subsequent transport occurred via the classical import pathway. The interaction between the N. crassa importin-α (NcImpα) and the NIT-2 NLS was quantified with calorimetric assays, leading to the observation that the peptide bound to two sites with different affinities, which is typical of a monopartite NLS sequence. The crystal structure of the NcImpα/NIT-2 NLS complex was solved and revealed that the NIT-2 peptide binds to NcImpα with the major NLS-binding site playing a primary role. This result contrasts other recent studies that suggested a major role for the minor NLS-binding site in importin-α from the α2 family, indicating that both sites can be used for different cargo proteins according to specific metabolic requirements.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas Fúngicas/metabolismo , Neurospora crassa/metabolismo , Fatores de Transcrição/metabolismo , alfa Carioferinas/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/fisiologia , Células Cultivadas , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Células HeLa , Humanos , Neurospora crassa/genética , Estrutura Secundária de Proteína , Esporos Fúngicos , Fatores de Transcrição/química , Fatores de Transcrição/genética , Difração de Raios X , alfa Carioferinas/química , alfa Carioferinas/genética
5.
J Cell Sci ; 128(15): 2759-65, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26065430

RESUMO

Gp78 (also known as AMFR), an endoplasmic-reticulum (ER)-associated protein degradation (ERAD) E3 ubiquitin ligase, localizes to mitochondria-associated ER and targets the mitofusin (Mfn1 and Mfn2) mitochondrial fusion proteins for degradation. Gp78 is also the cell surface receptor for autocrine motility factor (AMF), which prevents Gp78-dependent mitofusin degradation. Gp78 ubiquitin ligase activity promotes ER-mitochondria association and ER-mitochondria Ca(2+) coupling, processes that are reversed by AMF. Electron microscopy of HT-1080 fibrosarcoma cancer cells identified both smooth ER (SER; ∼8 nm) and wider (∼50-60 nm) rough ER (RER)-mitochondria contacts. Both short hairpin RNA (shRNA)-mediated knockdown of Gp78 (shGp78) and AMF treatment selectively reduced the extent of RER-mitochondria contacts without impacting on SER--mitochondria contacts. Concomitant small interfering RNA (siRNA)-mediated knockdown of Mfn1 increased SER-mitochondria contacts in both control and shGp78 cells, whereas knockdown of Mfn2 increased RER-mitochondria contacts selectively in shGp78 HT-1080 cells. The mitofusins therefore inhibit ER-mitochondria interaction. Regulation of close SER-mitochondria contacts by Mfn1 and of RER-mitochondria contacts by AMF-sensitive Gp78-mediated degradation of Mfn2 define new mechanisms that regulate ER-mitochondria interactions.


Assuntos
Retículo Endoplasmático Rugoso/genética , Retículo Endoplasmático Liso/genética , GTP Fosfo-Hidrolases/genética , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas Mitocondriais/genética , Receptores do Fator Autócrino de Motilidade/genética , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Retículo Endoplasmático Rugoso/metabolismo , Retículo Endoplasmático Liso/metabolismo , Degradação Associada com o Retículo Endoplasmático/fisiologia , Humanos , Mitocôndrias , Interferência de RNA , RNA Interferente Pequeno
6.
PLoS Pathog ; 9(10): e1003671, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204256

RESUMO

Disassembly of the nuclear lamina is essential in mitosis and apoptosis requiring multiple coordinated enzymatic activities in nucleus and cytoplasm. Activation and coordination of the different activities is poorly understood and moreover complicated as some factors translocate between cytoplasm and nucleus in preparatory phases. Here we used the ability of parvoviruses to induce nuclear membrane breakdown to understand the triggers of key mitotic enzymes. Nuclear envelope disintegration was shown upon infection, microinjection but also upon their application to permeabilized cells. The latter technique also showed that nuclear envelope disintegration was independent upon soluble cytoplasmic factors. Using time-lapse microscopy, we observed that nuclear disassembly exhibited mitosis-like kinetics and occurred suddenly, implying a catastrophic event irrespective of cell- or type of parvovirus used. Analyzing the order of the processes allowed us to propose a model starting with direct binding of parvoviruses to distinct proteins of the nuclear pore causing structural rearrangement of the parvoviruses. The resulting exposure of domains comprising amphipathic helices was required for nuclear envelope disintegration, which comprised disruption of inner and outer nuclear membrane as shown by electron microscopy. Consistent with Ca⁺⁺ efflux from the lumen between inner and outer nuclear membrane we found that Ca⁺⁺ was essential for nuclear disassembly by activating PKC. PKC activation then triggered activation of cdk-2, which became further activated by caspase-3. Collectively our study shows a unique interaction of a virus with the nuclear envelope, provides evidence that a nuclear pool of executing enzymes is sufficient for nuclear disassembly in quiescent cells, and demonstrates that nuclear disassembly can be uncoupled from initial phases of mitosis.


Assuntos
Sinalização do Cálcio , Caspase 3/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Parvovirus H-1/metabolismo , Mitose , Membrana Nuclear/enzimologia , Infecções por Parvoviridae/enzimologia , Proteína Quinase C/metabolismo , Animais , Cálcio/metabolismo , Caspase 3/genética , Quinase 2 Dependente de Ciclina/genética , Parvovirus H-1/genética , Células HeLa , Humanos , Membrana Nuclear/genética , Membrana Nuclear/patologia , Membrana Nuclear/virologia , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/patologia , Proteína Quinase C/genética , Xenopus laevis
7.
Viruses ; 15(8)2023 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-37631984

RESUMO

Replication of the RNA genome of influenza A virus occurs in the nucleus of infected cells. The influenza nucleoprotein (NP) associated with the viral RNA into ribonucleoprotein complexes (vRNPs) is involved in the nuclear import of the viral genome. NP has two nuclear localization sequences (NLSs), NLS1 and NLS2. Most studies have concentrated on the role of NP's NLSs using in vitro-assembled or purified vRNPs, which may differ from incoming vRNPs released in the cytoplasm during an infection. Here, we study the contribution of the NP's NLSs to the nuclear import of vRNPs in a cell culture model system for influenza infection: human lung carcinoma cells infected with viruses containing NP-carrying mutations in NLS1 or NLS2 (NLS2MT), generated by reverse genetics. We found that cells infected with these mutant viruses were defective in the nuclear import of incoming vRNPs and produced reduced amounts of newly synthesized NP, newly assembled vRNP, and progeny virus. In addition, NLS2MT-infected cells were also defective in the nucleolar accumulation of NP, confirming the nucleolar localization role of NLS2. Our findings indicate that both NLS1 and NLS2 have to be present for successful infection and demonstrate the crucial role of these two NLSs in the infection cycle of the influenza A virus.


Assuntos
Vírus da Influenza A , Influenza Humana , Humanos , Transporte Ativo do Núcleo Celular , Núcleo Celular , Vírus da Influenza A/genética , Nucleoproteínas/genética , RNA Viral/genética
8.
J Struct Biol ; 177(1): 90-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22100338

RESUMO

Baculoviruses are one of the largest viruses that replicate in the nucleus of their host cells. During an infection the capsid, containing the DNA viral genome, is released into the cytoplasm and delivers the genome into the nucleus by a mechanism that is largely unknown. Here, we used capsids of the baculovirus Autographa californica multiple nucleopolyhedrovirus in combination with electron microscopy and discovered this capsid crosses the NPC and enters into the nucleus intact, where it releases its genome. To better illustrate the existence of this capsid through the NPC in its native conformation, we reconstructed the nuclear import event using electron tomography. In addition, using different experimental conditions, we were able to visualize the intact capsid interacting with NPC cytoplasmic filaments, as an initial docking site, and midway through the NPC. Our data suggests the NPC central channel undergoes large-scale rearrangements to allow translocation of the intact 250-nm long baculovirus capsid. We discuss our results in the light of the hypothetical models of NPC function.


Assuntos
Transporte Ativo do Núcleo Celular , Baculoviridae/ultraestrutura , DNA Viral/química , Poro Nuclear/ultraestrutura , Animais , Baculoviridae/metabolismo , Capsídeo/química , Proteínas do Capsídeo/metabolismo , Citoplasma/metabolismo , DNA Viral/isolamento & purificação , Tomografia com Microscopia Eletrônica , Oócitos , Xenopus laevis
9.
Biochim Biophys Acta ; 1813(9): 1634-45, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21167871

RESUMO

Many viruses depend on nuclear proteins for replication. Therefore, their viral genome must enter the nucleus of the host cell. In this review we briefly summarize the principles of nucleocytoplasmic transport, and then describe the diverse strategies used by viruses to deliver their genomes into the host nucleus. Some of the emerging mechanisms include: (1) nuclear entry during mitosis, when the nuclear envelope is disassembled, (2) viral genome release in the cytoplasm followed by entry of the genome through the nuclear pore complex (NPC), (3) capsid docking at the cytoplasmic side of the NPC, followed by genome release, (4) nuclear entry of intact capsids through the NPC, followed by genome release, and (5) nuclear entry via virus-induced disruption of the nuclear envelope. Which mechanism a particular virus uses depends on the size and structure of the virus, as well as the cellular cues used by the virus to trigger capsid disassembly and genome release. This article is part of a Special Issue entitled: Regulation of Signaling and Cellular Fate through Modulation of Nuclear Protein Import.


Assuntos
Núcleo Celular/fisiologia , Fenômenos Fisiológicos Virais , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Genoma Viral , Humanos , Microscopia Eletrônica de Transmissão , Mitose/fisiologia , Modelos Biológicos , Membrana Nuclear/fisiologia , Membrana Nuclear/ultraestrutura , Membrana Nuclear/virologia , Poro Nuclear/fisiologia , Poro Nuclear/ultraestrutura , Poro Nuclear/virologia , Internalização do Vírus , Vírus/genética , Vírus/ultraestrutura
10.
J Virol ; 85(10): 4863-74, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21367902

RESUMO

Parvoviruses are small, nonenveloped, single-stranded DNA viruses which replicate in the nucleus of the host cell. We have previously found that early during infection the parvovirus minute virus of mice (MVM) causes small, transient disruptions of the nuclear envelope (NE). We have now investigated the mechanism used by MVM to disrupt the NE. Here we show that the viral phospholipase A2, the only known enzymatic domain on the parvovirus capsid, is not involved in causing NE disruption. Instead, the virus utilizes host cell caspases, which are proteases involved in causing NE breakdown during apoptosis, to facilitate these nuclear membrane disruptions. Studies with pharmacological inhibitors indicate that caspase-3 in particular is involved. A caspase-3 inhibitor prevents nuclear lamin cleavage and NE disruption in MVM-infected mouse fibroblast cells and reduces nuclear entry of MVM capsids and viral gene expression. Caspase-3 is, however, not activated above basal levels in MVM-infected cells, and other aspects of apoptosis are not triggered during early MVM infection. Instead, basally active caspase-3 is relocalized to the nuclei of infected cells. We propose that NE disruption involving caspases plays a role in (i) parvovirus entry into the nucleus and (ii) alteration of the compartmentalization of host proteins in a way that is favorable for the virus.


Assuntos
Caspase 3/metabolismo , Vírus Miúdo do Camundongo/patogenicidade , Membrana Nuclear/patologia , Animais , Linhagem Celular , Fibroblastos/virologia , Humanos , Camundongos , Fosfolipases A2/metabolismo , Proteínas Virais/metabolismo
11.
PLoS Pathog ; 6(1): e1000741, 2010 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-20126445

RESUMO

Virtually all DNA viruses including hepatitis B viruses (HBV) replicate their genome inside the nucleus. In non-dividing cells, the genome has to pass through the nuclear pore complexes (NPCs) by the aid of nuclear transport receptors as e.g. importin beta (karyopherin). Most viruses release their genome in the cytoplasm or at the cytosolic face of the NPC, as the diameter of their capsids exceeds the size of the NPC. The DNA genome of HBV is derived from reverse transcription of an RNA pregenome. Genome maturation occurs in cytosolic capsids and progeny capsids can deliver the genome into the nucleus causing nuclear genome amplification. The karyophilic capsids are small enough to pass the NPC, but nuclear entry of capsids with an immature genome is halted in the nuclear basket on the nuclear side of the NPC, and the genome remains encapsidated. In contrast, capsids with a mature genome enter the basket and consequently liberate the genome. Investigating the difference between immature and mature capsids, we found that mature capsids had to disintegrate in order to leave the nuclear basket. The arrest of a karyophilic cargo at the nuclear pore is a rare phenomenon, which has been described for only very few cellular proteins participating in nuclear entry. We analyzed the interactions causing HBV capsid retention. By pull-down assays and partial siRNA depletion, we showed that HBV capsids directly interact with nucleoporin 153 (Nup153), an essential protein of the nuclear basket which participates in nuclear transport via importin beta. The binding sites of importin beta and capsids were shown to overlap but capsid binding was 150-fold stronger. In cellulo experiments using digitonin-permeabilized cells confirmed the interference between capsid binding and nuclear import by importin beta. Collectively, our findings describe a unique nuclear import strategy not only for viruses but for all karyophilic cargos.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Proteínas do Capsídeo/metabolismo , Núcleo Celular/metabolismo , Vírus da Hepatite B/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Replicação Viral/fisiologia , Animais , Núcleo Celular/virologia , Células HeLa , Humanos , Imunoprecipitação , RNA Interferente Pequeno , Xenopus laevis
12.
J Cell Biol ; 178(5): 785-98, 2007 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-17724119

RESUMO

Sun1 and 2 are A-type lamin-binding proteins that, in association with nesprins, form a link between the inner nuclear membranes (INMs) and outer nuclear membranes of mammalian nuclear envelopes. Both immunofluorescence and immunoelectron microscopy reveal that Sun1 but not Sun2 is intimately associated with nuclear pore complexes (NPCs). Topological analyses indicate that Sun1 is a type II integral protein of the INM. Localization of Sun1 to the INM is defined by at least two discrete regions within its nucleoplasmic domain. However, association with NPCs is dependent on the synergy of both nucleoplasmic and lumenal domains. Cells that are either depleted of Sun1 by RNA interference or that overexpress dominant-negative Sun1 fragments exhibit clustering of NPCs. The implication is that Sun1 represents an important determinant of NPC distribution across the nuclear surface.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Poro Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Motivos de Aminoácidos , Animais , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas Associadas aos Microtúbulos/química , Proteínas Associadas aos Microtúbulos/genética , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Estrutura Quaternária de Proteína , Interferência de RNA , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
13.
Cells ; 11(19)2022 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-36230922

RESUMO

Influenza viruses deliver their genome into the nucleus of infected cells for replication. This process is mediated by the viral nucleoprotein (NP), which contains two nuclear localization sequences (NLSs): NLS1 at the N-terminus and a recently identified NLS2 (212GRKTR216). Through mutagenesis and functional studies, we demonstrated that NP must have both NLSs for an efficient nuclear import. As with other NLSs, there may be variations in the basic residues of NLS2 in different strains of the virus, which may affect the nuclear import of the viral genome. Although all NLS2 variants fused to the GFP mediated nuclear import of GFP, bioinformatics showed that 98.8% of reported NP sequences contained either the wild-type sequence 212GRKTR216 or 212GRRTR216. Bioinformatics analyses used to study the presence of NLS2 variants in other viral and nuclear proteins resulted in very low hits, with only 0.4% of human nuclear proteins containing putative NLS2. From these, we studied the nucleolar protein 14 (NOP14) and found that NLS2 does not play a role in the nuclear import of this protein but in its nucleolar localization. We also discovered a functional NLS at the C-terminus of NOP14. Our findings indicate that NLS2 is a highly conserved influenza A NP sequence.


Assuntos
Biologia Computacional , Sinais de Localização Nuclear , Sequência de Aminoácidos , Humanos , Sinais de Localização Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Proteínas do Nucleocapsídeo , Nucleoproteínas/metabolismo
14.
J Biol Chem ; 285(18): 13769-80, 2010 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-20197273

RESUMO

Importin beta mediates active passage of cellular substrates through the nuclear pore complex (NPC). Adaptors such as importin alpha and snurportin associate with importin beta via an importin beta binding (IBB) domain. The intrinsic structural flexibility of importin beta allows its concerted interactions with IBB domains, phenylalanine-glycine nucleoporins, and the GTPase Ran during transport. In this paper, we provide evidence that the nature of the IBB domain modulates the affinity of the import complex for the NPC. In permeabilized cells, importin beta imports a cargo fused to the snurportin IBB (sIBB) with approximately 70% reduced energy requirement as compared with the classical importin alpha IBB. At the molecular level, this is explained by approximately 200-fold reduced affinity of importin beta for Nup62, when bound to the sIBB. Consistently, in vivo, the importin beta.sIBB complex has greatly reduced persistence inside the central channel of the NPC. We propose that by controlling the degree of strain in the tertiary structure of importin beta, the IBB domain modulates the affinity of the import complex for nucleoporins, thus dictating its persistence inside the NPC.


Assuntos
Poro Nuclear/metabolismo , beta Carioferinas/metabolismo , Células HeLa , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Poro Nuclear/genética , Complexo de Proteínas Formadoras de Poros Nucleares , Estrutura Terciária de Proteína , Proteínas de Ligação ao Cap de RNA/genética , Proteínas de Ligação ao Cap de RNA/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , alfa Carioferinas/genética , alfa Carioferinas/metabolismo , beta Carioferinas/genética , Proteína ran de Ligação ao GTP/genética , Proteína ran de Ligação ao GTP/metabolismo
15.
Methods ; 51(1): 114-20, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20138149

RESUMO

Microinjection of Xenopus laevis oocytes is an excellent system for studying nuclear transport because of the large size of the oocyte and its high nuclear pore complex (NPC) density. In addition, the fact that Xenopus oocytes are not permissive for most mammalian viruses makes this system especially useful for studying nuclear transport of viruses in the absence of the confounding factor of virus replication. In this article, we briefly discuss the contribution of microinjection of Xenopus oocytes to the field of nuclear transport. We then describe the protocols we have developed using microinjection of Xenopus oocytes to study nuclear transport of viral capsids, and summarize variations of the technique that can be used to address many different questions about the nuclear transport of viruses.


Assuntos
Transporte Ativo do Núcleo Celular , Oócitos/citologia , Vírus/metabolismo , Xenopus laevis/metabolismo , Animais , Western Blotting , Capsídeo/metabolismo , Imuno-Histoquímica , Microinjeções , Microscopia Eletrônica/métodos , Modelos Biológicos , Membrana Nuclear/virologia , Oócitos/metabolismo , Parvovirus/metabolismo , Vírus/química , Xenopus/metabolismo , Xenopus laevis/virologia
16.
Dev Cell ; 7(6): 780-1, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15572121

RESUMO

Nuclear pore complexes (NPCs) are large protein assemblies embedded in the nuclear envelope that act as passageways for transport of molecules into and out of the nucleus. Two new studies, one in Nature Cell Biology and one in Science , offer direct evidence that the NPC is a highly dynamic structure.


Assuntos
Complexo de Proteínas Formadoras de Poros Nucleares/química , Poro Nuclear/fisiologia , Animais , Anisotropia , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Citoplasma/metabolismo , Dictyostelium , Proteínas de Fluorescência Verde/metabolismo , Humanos , Microscopia Eletrônica , Modelos Biológicos , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo
17.
Virol J ; 6: 68, 2009 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-19490630

RESUMO

BACKGROUND: Early in infection, the genome of the influenza A virus, consisting of eight complexes of RNA and proteins (termed viral ribonucleoproteins; vRNPs), enters the nucleus of infected cells for replication. Incoming vRNPs are imported into the nucleus of infected cells using at least two nuclear localization sequences on nucleoprotein (NP; NLS1 at the N terminus, and NLS2 in the middle of the protein). Progeny vRNP assembly occurs in the nucleus, and later in infection, these are exported from the nucleus to the cytoplasm. Nuclear-exported vRNPs are different from incoming vRNPs in that they are prevented from re-entering the nucleus. Why nuclear-exported vRNPs do not re-enter the nucleus is unknown. RESULTS: To test our hypothesis that the exposure of NLSs on the vRNP regulates the directionality of the nuclear transport of the influenza vRNPs, we immunolabeled the two NLSs of NP (NLS1 and NLS2) and analyzed their surface accessibility in cells infected with the influenza A virus. We found that the NLS1 epitope on NP was exposed throughout the infected cells, but the NLS2 epitope on NP was only exposed in the nucleus of the infected cells. Addition of the nuclear export inhibitor leptomycin B further revealed that NLS1 is no longer exposed in cytoplasmic NP and vRNPs that have already undergone nuclear export. Similar immunolabeling studies in the presence of leptomycin B and with cells transfected with the cDNA of NP revealed that the NLS1 on NP is hidden in nuclear exported-NP. CONCLUSION: NLS1 mediates the nuclear import of newly-synthesized NP and incoming vRNPs. This NLS becomes hidden on nuclear-exported NP and nuclear-exported vRNPs. Thus the selective exposure of the NLS1 constitutes a critical mechanism to regulate the directionality of the nuclear transport of vRNPs during the influenza A viral life cycle.


Assuntos
Núcleo Celular/metabolismo , Vírus da Influenza A/fisiologia , Sinais Direcionadores de Proteínas , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/metabolismo , Replicação Viral , Transporte Ativo do Núcleo Celular , Núcleo Celular/virologia , Células HeLa , Humanos , Proteínas do Nucleocapsídeo
18.
PLoS One ; 14(8): e0221562, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31437229

RESUMO

The transport of macromolecules into the cell nucleus occurs through nuclear pore complexes (NPCs) and is mediated by cellular receptors. Recently, a novel mechanism of nuclear entry, in which actin polymerization provides a propulsive force driving the transport through the NPC, has been proposed. This mechanism is used by the nucleocapsid from baculovirus, one of the largest viruses to replicate in the nucleus of their host cells, which crosses the NPC and enters the nucleus independently of cellular receptors. The baculovirus nucleocapsid contains a protein that hijacks the cellular actin polymerization machinery to assemble actin filaments that propel the nucleocapsid through the host cell cytoplasm. In this study, we functionalized carbon nanotubes by covalently attaching a protein domain responsible for inducing actin polymerization and investigated their nuclear entry. We found that the functionalized carbon nanotubes were able to enter the cell nucleus under permissive conditions for actin polymerization, but not when this process was inhibited. We conclude that the mechanical force generated by actin polymerization can drive cargo entry into the cell nucleus. Our results support a novel force-driven mechanism for molecular entry into the cell nucleus.


Assuntos
Actinas/metabolismo , Núcleo Celular/metabolismo , Nanotubos de Carbono/química , Polimerização , Complexo 2-3 de Proteínas Relacionadas à Actina/química , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Bovinos , Permeabilidade da Membrana Celular , Células HeLa , Humanos , Nanotubos de Carbono/ultraestrutura , Domínios Proteicos , Soroalbumina Bovina/química
19.
FEBS Open Bio ; 9(7): 1174-1183, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31144423

RESUMO

The tumor suppressor activity of maspin (mammary serine protease inhibitor) has been associated with its nuclear localization. In this study we explore the regulation of maspin nuclear translocation. An in vitro nuclear import assay suggested that maspin can passively enter the nucleus. However, in silico analysis identified a putative maspin nuclear localization signal (NLS), which was able to mediate the nuclear translocation of a chimeric protein containing this NLS fused to five green fluorescent protein molecules in tandem (5GFP). Dominant-negative Ran-GTPase mutants RanQ69L or RanT24N suppressed this process. Unexpectedly, the full-length maspin fused to 5GFP failed to enter the nucleus. As maspin's putative NLS is partially hidden in its three-dimensional structure, we suggest that maspin nuclear transport could be conformationally regulated. Our results suggest that maspin nuclear translocation involves both passive and active mechanisms.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Sinais de Localização Nuclear/metabolismo , Serpinas/metabolismo , Núcleo Celular/metabolismo , Proteínas de Fluorescência Verde , Células HeLa , Humanos , Sinais de Localização Nuclear/fisiologia , Inibidores de Serina Proteinase/metabolismo , Serpinas/fisiologia , Serpinas/ultraestrutura , Proteína ran de Ligação ao GTP/metabolismo
20.
Mol Cancer Res ; 16(1): 16-31, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28993511

RESUMO

Cell-cycle progression and the acquisition of a migratory phenotype are hallmarks of human carcinoma cells that are perceived as independent processes but may be interconnected by molecular pathways that control microtubule nucleation at centrosomes. Here, cell-cycle progression dramatically impacts the engraftment kinetics of 4T1-luciferase2 breast cancer cells in immunocompetent BALB/c or immunocompromised NOD-SCID gamma (NSG) mice. Multiparameter imaging of wound closure assays was used to track cell-cycle progression, cell migration, and associated phenotypes in epithelial cells or carcinoma cells expressing a fluorescence ubiquitin cell-cycle indicator. Cell migration occurred with an elevated velocity and directionality during the S-G2-phase of the cell cycle, and cells in this phase possess front-polarized centrosomes with augmented microtubule nucleation capacity. Inhibition of Aurora kinase-A (AURKA/Aurora-A) dampens these phenotypes without altering cell-cycle progression. During G2-phase, the level of phosphorylated Aurora-A at centrosomes is reduced in hyaluronan-mediated motility receptor (HMMR)-silenced cells as is the nuclear transport of TPX2, an Aurora-A-activating protein. TPX2 nuclear transport depends upon HMMR-T703, which releases TPX2 from a complex with importin-α (KPNA2) at the nuclear envelope. Finally, the abundance of phosphorylated HMMR-T703, a substrate for Aurora-A, predicts breast cancer-specific survival and relapse-free survival in patients with estrogen receptor (ER)-negative (n = 941), triple-negative (TNBC) phenotype (n = 538), or basal-like subtype (n = 293) breast cancers, but not in those patients with ER-positive breast cancer (n = 2,218). Together, these data demonstrate an Aurora-A/TPX2/HMMR molecular axis that intersects cell-cycle progression and cell migration.Implications: Tumor cell engraftment, migration, and cell-cycle progression share common regulation of the microtubule cytoskeleton through the Aurora-A/TPX2/HMMR axis, which has the potential to influence the survival of patients with ER-negative breast tumors. Mol Cancer Res; 16(1); 16-31. ©2017 AACR.


Assuntos
Aurora Quinase A/genética , Proteínas de Ciclo Celular/metabolismo , Animais , Aurora Quinase A/metabolismo , Feminino , Humanos , Camundongos , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA