Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Sci Rep ; 7(1): 11381, 2017 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-28900157

RESUMO

The influenza A virus nucleoprotein (NP) is an essential multifunctional protein that encapsidates the viral genome and functions as an adapter between the virus and the host cell machinery. NPs from all strains of influenza A viruses contain two nuclear localization signals (NLSs): a well-studied monopartite NLS1 and a less-characterized NLS2, thought to be bipartite. Through site-directed mutagenesis and functional analysis, we found that NLS2 is also monopartite and is indispensable for viral infection. Atomic structures of importin α bound to two variants of NLS2 revealed NLS2 primarily binds the major-NLS binding site of importin α, unlike NLS1 that associates with the minor NLS-pocket. Though peptides corresponding to NLS1 and NLS2 bind weakly to importin α, the two NLSs synergize in the context of the full length NP to confer high avidity for importin α7, explaining why the virus efficiently replicates in the respiratory tract that exhibits high levels of this isoform. This study, the first to functionally characterize NLS2, demonstrates NLS2 plays an important and unexpected role in influenza A virus infection. We propose NLS1 and NLS2 form a bipartite NLS in trans, which ensures high avidity for importin α7 while preventing non-specific binding to viral RNA.


Assuntos
Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , Sinais de Localização Nuclear , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/genética , Proteínas do Core Viral/metabolismo , alfa Carioferinas/genética , alfa Carioferinas/metabolismo , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Linhagem Celular , Humanos , Influenza Humana/metabolismo , Influenza Humana/virologia , Modelos Moleculares , Conformação Molecular , Mutação , Proteínas do Nucleocapsídeo , Ligação Proteica , Proteínas de Ligação a RNA/química , Proteínas do Core Viral/química
10.
Sci Rep ; 7(1): 5515, 2017 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-28710431

RESUMO

HIV-1 co-opts several host machinery to generate a permissive environment for viral replication and transmission. In this work we reveal how HIV-1 impacts the host translation and intracellular vesicular trafficking machineries for protein synthesis and to impede the physiological late endosome/lysosome (LEL) trafficking in stressful conditions. First, HIV-1 enhances the activity of the master regulator of protein synthesis, the mammalian target of rapamycin (mTOR). Second, the virus commandeers mTOR-associated late endosome/lysosome (LEL) trafficking and counteracts metabolic and environmental stress-induced intracellular repositioning of LEL. We then show that the small Rag GTPases, RagA and RagB, are required for the HIV-1-mediated LEL repositioning that is likely mediated by interactions between the Rags and the viral proteins, Gag and Vif. siRNA-mediated depletion of RagA and RagB leads to a loss in mTOR association to LEL and to a blockade of viral particle assembly and release at the plasma membrane with a marked concomitant reduction in virus production. These results show that HIV-1 co-opts fundamental mechanisms that regulate LEL motility and positioning and support the notion that LEL positioning is critical for HIV-1 replication.


Assuntos
HIV-1/fisiologia , Lisossomos/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Células HeLa , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Monoméricas de Ligação ao GTP/genética , RNA Interferente Pequeno/farmacologia , Células THP-1 , Montagem de Vírus , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo
11.
Virology ; 497: 41-52, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27423069

RESUMO

Influenza A virus exploits the subcellular transport machinery during the early stages of infection. Actin filaments and microtubules facilitate the trafficking of virus-containing endosomes towards the perinuclear region; however, the role of vimentin remains to be determined. In this study, we followed influenza A virus infection in vimentin-null cells and found that vimentin depletion severely reduced influenza viral RNA and protein expression, and production of infectious progeny virions. Furthermore, we show that in vimentin-null cells endosomal distribution and acidification were affected, and incoming influenza virions accumulated in late endosomes of these cells. We propose that this accumulation resulted from the impaired acidification of late endosomes in vimentin-null cells, which blocked the release of the viral genome from these organelles. These findings are the first to demonstrate that vimentin is critical for influenza viral infection as it facilitates endosomal trafficking and acidification, and mediates viral genome penetration into the cytoplasm to propagate the infection.


Assuntos
Endossomos/metabolismo , Endossomos/virologia , Genoma Viral , Vírus da Influenza A/fisiologia , RNA Viral/genética , RNA Viral/metabolismo , Vimentina/metabolismo , Replicação Viral , Células Cultivadas , Expressão Gênica , Técnicas de Inativação de Genes , Genes Reporter , Humanos , Concentração de Íons de Hidrogênio , Influenza Humana/virologia , Lisossomos/metabolismo , Lisossomos/virologia , Pinocitose , Vimentina/genética
12.
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
13.
Front Microbiol ; 6: 467, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26029198

RESUMO

DNA viruses undertake their replication within the cell nucleus, and therefore they must first deliver their genome into the nucleus of their host cells. Thus, trafficking across the nuclear envelope is at the basis of DNA virus infections. Nuclear transport of molecules with diameters up to 39 nm is a tightly regulated process that occurs through the nuclear pore complex (NPC). Due to the enormous diversity of virus size and structure, each virus has developed its own strategy for entering the nucleus of their host cells, with no two strategies alike. For example, baculoviruses target their DNA-containing capsid to the NPC and subsequently enter the nucleus intact, while the hepatitis B virus capsid crosses the NPC but disassembles at the nuclear side of the NPC. For other viruses such as herpes simplex virus and adenovirus, although both dock at the NPC, they have each developed a distinct mechanism for the subsequent delivery of their genome into the nucleus. Remarkably, other DNA viruses, such as parvoviruses and human papillomaviruses, access the nucleus through an NPC-independent mechanism. This review discusses our current understanding of the mechanisms used by DNA viruses to deliver their genome into the nucleus, and further presents the experimental evidence for such mechanisms.

14.
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
15.
Virology ; 482: 157-66, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25863880

RESUMO

The minute virus of mice, prototype strain (MVMp), is a non-enveloped, single-stranded DNA virus of the family Parvoviridae. Unlike other parvoviruses, the mechanism of cellular uptake of MVMp has not been studied in detail. We analyzed MVMp endocytosis in mouse LA9 fibroblasts and a tumor cell line derived from epithelial-mesenchymal transition through polyomavirus middle T antigen transformation in transgenic mice. By a combination of immunofluorescence and electron microscopy, we found that MVMp endocytosis occurs at the leading edge of migrating cells in proximity to focal adhesion sites. By using drug inhibitors of various endocytic pathways together with immunofluorescence microscopy and flow cytometry analysis, we discovered that MVMp can use a number of endocytic pathways, depending on the host cell type. At least three different mechanisms were identified: clathrin-, caveolin-, and clathrin-independent carrier-mediated endocytosis, with the latter occurring in transformed cells but not in LA9 fibroblasts.


Assuntos
Endocitose , Vírus Miúdo do Camundongo/fisiologia , Internalização do Vírus , Animais , Linhagem Celular , Células Epiteliais/virologia , Fibroblastos/virologia , Citometria de Fluxo , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Microscopia de Fluorescência
16.
Curr Opin Virol ; 12: 59-65, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25846849

RESUMO

The nuclear import of viral genomes is an important step of the infectious cycle for viruses that replicate in the nucleus of their host cells. Although most viruses use the cellular nuclear import machinery or some components of this machinery, others have developed sophisticated ways to reach the nucleus. Some of these have been known for some time; however, recent studies have changed our understanding of how some non-enveloped DNA viruses access the nucleus. For example, parvoviruses enter the nucleus through small disruptions of the nuclear membranes and nuclear lamina, and adenovirus tugs at the nuclear pore complex, using kinesin-1, to disassemble their capsids and deliver viral proteins and genomes into the nucleus. Here we review recent findings of the nuclear import strategies of three small non-enveloped DNA viruses, including adenovirus, parvovirus, and the polyomavirus simian virus 40.


Assuntos
Núcleo Celular/metabolismo , Núcleo Celular/virologia , Vírus de DNA/fisiologia , DNA Viral/metabolismo , Poro Nuclear/virologia , Transporte Ativo do Núcleo Celular , Adenoviridae/fisiologia , Vírus de DNA/genética , Genoma Viral , Humanos , Parvovirus/fisiologia , Vírus 40 dos Símios/fisiologia , Internalização do Vírus , Replicação Viral
17.
Virology ; 481: 63-72, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25768892

RESUMO

Galectin-3 has previously been found to be required by the parvovirus minute virus of mice prototype strain (MVMp) for infection of mouse fibroblast cells. Since MVMp is an oncotropic virus, and galectin-3 is a multifunctional protein implicated in cancer metastasis, we hypothesized that galectin-3 and Mgat5, the Golgi enzyme that synthesizes high-affinity glycan ligands of galectin-3, might play a role in MVMp infection. Using siRNA-mediated knockdown of galectin-3 in mouse cells transformed with polyomavirus middle T antigen and Mgat5(-/-) mouse mammary tumor cells, we found that galectin-3 and Mgat5 are both necessary for efficient MVMp cell entry and infection, but not for cell binding. Moreover, we found that human cancer cells expressing higher levels of galectin-3 were more efficiently infected with MVMp than cell lines expressing lower galectin-3 levels. We conclude that galectin-3 and Mgat5 are involved in MVMp infection, and propose that galectin-3 is a determinant of MVMp oncotropism.


Assuntos
Galectina 3/metabolismo , Vírus Miúdo do Camundongo/fisiologia , Infecções por Parvoviridae/veterinária , Doenças dos Roedores/metabolismo , Animais , Linhagem Celular , Galectina 3/genética , Humanos , Camundongos , Vírus Miúdo do Camundongo/genética , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Infecções por Parvoviridae/genética , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Doenças dos Roedores/genética , Doenças dos Roedores/virologia
18.
Virology ; 468-470: 150-159, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25173091

RESUMO

The parvovirus minute virus of mice, prototype strain (MVMp), preferentially infects and kills cancer cells. This intrinsic MVMp oncotropism may depend in part on the early stages of MVMp infection. To test this hypothesis, we investigated the early events of MVMp infection in mouse LA9 fibroblasts and a highly invasive mouse mammary tumor cell line derived from polyomavirus middle T antigen-mediated transformation. Using a combination of fluorescence and electron microscopy, we found that various parameters of the cell migration process affect MVMp infection. We show that, after binding to the plasma membrane, MVMp particles rapidly cluster at the leading edge of migrating cells, which exhibit higher levels of MVMp uptake than non-motile cells. Moreover, promoting cell migration on a fibronectin matrix increased MVMp infection, and induction of epithelial-mesenchymal transition allowed MVMp replication in non-permissive epithelial cells. Hence, we propose that cell migration influences the early stages of MVMp infection.


Assuntos
Movimento Celular/fisiologia , Vírus Miúdo do Camundongo/fisiologia , Animais , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Células Epiteliais/virologia , Transição Epitelial-Mesenquimal , Fibronectinas , Camundongos
19.
Methods Cell Biol ; 122: 81-98, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24857726

RESUMO

Xenopus oocytes are large in size and perfectly suited for microinjection experiments. Their nuclei, which can be readily isolated manually, are characterized by an extremely high density of nuclear pore complexes (NPCs). Therefore, Xenopus oocytes are an excellent system to study NPC structure and molecular architecture, as well as nucleocytoplasmic transport on an ultrastructural level. A wide range of electron microscopy (EM) techniques can be employed to do so and thin-sectioning immuno-EM has been proven to be a powerful tool in this context. NPCs are composed of multiple copies of a set of about 30 different nucleoporins, which are often large, multidomain proteins. Their complex organization within NPCs can be unraveled by using domain-specific antibodies to individual nucleoporins in combination with microinjection and expression of epitope-tagged nucleoporins. Here, we describe the immuno-EM methods using Xenopus oocyte that allow for precise ultrastructural localization of nucleoporins within the structure of the NPC.


Assuntos
Microscopia Imunoeletrônica/métodos , Complexo de Proteínas Formadoras de Poros Nucleares/imunologia , Poro Nuclear/química , Oócitos/citologia , Transporte Ativo do Núcleo Celular , Animais , Anticorpos Monoclonais/imunologia , Epitopos/imunologia , Feminino , Coloide de Ouro , Microinjeções/métodos , Microtomia , Estrutura Terciária de Proteína , Coloração e Rotulagem , Xenopus laevis
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...