Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biotechnol Adv ; 71: 108322, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38336188

RESUMEN

Recombinant adeno-associated virus (rAAV) is one of the prominent gene delivery vehicles that has opened promising opportunities for novel gene therapeutic approaches. However, the current major viral vector production platform, triple transfection in mammalian cells, may not meet the increasing demand. Thus, it is highly required to understand production bottlenecks from the host cell perspective and engineer the cells to be more favorable and tolerant to viral vector production, thereby effectively enhancing rAAV manufacturing. In this review, we provided a comprehensive exploration of the intricate cellular process involved in rAAV production, encompassing various stages such as plasmid entry to the cytoplasm, plasmid trafficking and nuclear delivery, rAAV structural/non-structural protein expression, viral capsid assembly, genome replication, genome packaging, and rAAV release/secretion. The knowledge in the fundamental biology of host cells supporting viral replication as manufacturing factories or exhibiting defending behaviors against viral production is summarized for each stage. The control strategies from the perspectives of host cell and materials (e.g., AAV plasmids) are proposed as our insights based on the characterization of molecular features and our existing knowledge of the AAV viral life cycle, rAAV and other viral vector production in the Human embryonic kidney (HEK) cells.


Asunto(s)
Dependovirus , Mamíferos , Humanos , Animales , Dependovirus/genética , Citoplasma , Transfección
2.
Biochim Biophys Acta Mol Cell Res ; 1871(3): 119685, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38342311

RESUMEN

The best-known function of the essential GPN-loop GTPase Gpn3 is to contribute to RNA polymerase II assembly, a prerequisite for its nuclear targeting. Although this process occurs in the cytoplasm, we have previously shown that Gpn3 enters the cell nucleus before being polyubiquitinated. Here, we show that inhibiting Crm1-mediated nuclear export with leptomycin B, or the proteasome with MG132, caused the nuclear accumulation of recombinant and endogenous Gpn3 in MCF-12A cells. When added simultaneously, leptomycin B and MG132 had an additive effect. Analysis of Gpn3 primary sequence revealed the presence of at least five nuclear export sequence (NES) motifs, with some having a higher exposure to the solvent in the GTP-bound than GDP-bound state in a Gpn3 structural model. Inactivation of any of these NESes led to some degree of Gpn3 nuclear accumulation, although mutating NES1 or NES3 had the more robust effect. MCF-12A cells expressing exclusively a NES-deficient version of Gpn3R-Flag proliferated slower than cells expressing Gpn3R-Flag wt, indicating that nuclear export is important for Gpn3 function. Next, we searched for physiological conditions regulating Gpn3 nucleocytoplasmic shuttling. Interestingly, whereas Gpn3R-Flag was both nuclear and cytoplasmic in low-density growing MCF-12A cells, it was exclusively cytoplasmic in high-density areas. Furthermore, Gpn3R-Flag was cytoplasmic, mostly perinuclear, in sparse but starved MCF-12A cells, and serum-stimulation caused a rapid, although transient, Gpn3R-Flag nuclear accumulation. We conclude that Gpn3 nucleocytoplasmic shuttling is regulated by cell density and growth factors, and propose that Gpn3 has an unknown nuclear function positively linked to cell growth and/or proliferation.


Asunto(s)
Núcleo Celular , GTP Fosfohidrolasas , GTP Fosfohidrolasas/metabolismo , Transporte Activo de Núcleo Celular , Núcleo Celular/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Recuento de Células
3.
mBio ; 15(1): e0021222, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38085100

RESUMEN

IMPORTANCE: HIV-1 capsid protein (CA)-independently or by recruiting host factors-mediates several key steps of virus replication in the cytoplasm and nucleus of the target cell. Research in the recent years have established that CA is multifunctional and genetically fragile of all the HIV-1 proteins. Accordingly, CA has emerged as a validated and high priority therapeutic target, and the first CA-targeting antiviral drug was recently approved for treating multi-drug resistant HIV-1 infection. However, development of next generation CA inhibitors depends on a better understanding of CA's known roles, as well as probing of CA's novel roles, in HIV-1 replication. In this timely review, we present an updated overview of the current state of our understanding of CA's multifunctional role in HIV-1 replication-with a special emphasis on CA's newfound post-nuclear roles, highlight the pressing knowledge gaps, and discuss directions for future research.


Asunto(s)
Infecciones por VIH , Seropositividad para VIH , VIH-1 , Humanos , Cápside/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , VIH-1/genética , VIH-1/metabolismo , Seropositividad para VIH/metabolismo , Replicación Viral/genética , Integración Viral
4.
Cell Mol Bioeng ; 16(5-6): 459-474, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38099212

RESUMEN

Introduction: Plasmid DNA (pDNA) must be delivered into the nucleus for transgene expression in mammalian cells. The entry may happen passively during the nuclear envelope breakdown and reformation in dividing cells or actively through the nuclear pore complexes. The goal of this study was to investigate the relative importance of these two pathways for pDNA nuclear entry and subsequent gene expression. Methods: To measure nuclear entry of pDNA encoding enhanced green florescence protein (EGFP) in electrotransfected cells, we developed a sensitive technique for quantitative analysis of pDNA in the nuclei, based on a hybridization probe for pDNA detection at the single molecule level and automatic image analysis. In matched experiments, we used an mRNA targeted hybridization probe to quantify reporter mRNA expression per cell, and flow cytometry to quantify expression of EGFP. Results: We discovered two distinct patterns of pDNA distribution in the nuclei: punctate and diffuse, which were dominant in arrested and unarrested cells, respectively. The cell cycle arrest decreased diffuse pDNA and increased punctate pDNA. Its net effect was a decrease in the total intranuclear pDNA. Additionally, the cell cycle arrest increased the reporter mRNA synthesis but had no substantial impact on reporter protein expression. Conclusion: Results from the study demonstrated that the efficient nuclear entry of pDNA during cell division did not necessarily lead to a high level of transgene expression. They also suggested that the punctate pDNA was more transcriptionally active than diffuse pDNA in the nuclei. These data will be useful in future studies for understanding mechanisms of nonviral gene delivery.

5.
J Virol ; 97(11): e0073223, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37843371

RESUMEN

The HIV-1 genome encodes a small number of proteins with structural, enzymatic, regulatory, and accessory functions. These viral proteins interact with a number of host factors to promote the early and late stages of HIV-1 infection. During the early stages of infection, interactions between the viral proteins and host factors enable HIV-1 to enter the target cell, traverse the cytosol, dock at the nuclear pore, gain access to the nucleus, and integrate into the host genome. Similarly, the viral proteins recruit another set of host factors during the late stages of infection to orchestrate HIV-1 transcription, translation, assembly, and release of progeny virions. Among the host factors implicated in HIV-1 infection, Cyclophilin A (CypA) was identified as the first host factor to be packaged within HIV-1 particles. It is now well established that CypA promotes HIV-1 infection by directly binding to the viral capsid. Mechanistic models to pinpoint CypA's role have spanned from an effect in the producer cell to the early steps of infection in the target cell. In this review, we will describe our understanding of the role(s) of CypA in HIV-1 infection, highlight the current knowledge gaps, and discuss the potential role of this host factor in the post-nuclear entry steps of HIV-1 infection.


Asunto(s)
Ciclofilina A , Infecciones por VIH , VIH-1 , Humanos , Proteínas de la Cápside/genética , Núcleo Celular/metabolismo , Ciclofilina A/genética , Ciclofilina A/metabolismo , Infecciones por VIH/metabolismo , VIH-1/genética , VIH-1/metabolismo , Proteínas Virales/metabolismo , Interacciones Huésped-Patógeno
6.
J Virol Methods ; 322: 114834, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37875225

RESUMEN

HIV-1 enters the nucleus of non-dividing cells through the nuclear pore complex where it integrates into the host genome. The mechanism of HIV-1 nuclear import remains poorly understood. A powerful means to investigate the docking of HIV-1 at the nuclear pore and nuclear import of viral complexes is through single virus tracking in live cells. This approach necessitates fluorescence labeling of HIV-1 particles and the nuclear envelope, which may be challenging, especially in the context of primary cells. Here, we leveraged a deep neural network model for label-free visualization of the nuclear envelope using transmitted light microscopy. A training image set of cells with fluorescently labeled nuclear Lamin B1 (ground truth), along with the corresponding transmitted light images, was acquired and used to train our model to predict the morphology of the nuclear envelope in fixed cells. This protocol yielded accurate predictions of the nuclear membrane and was used in conjunction with virus infection to examine the nuclear entry of fluorescently labeled HIV-1 complexes. Analyses of HIV-1 nuclear import as a function of virus input yielded identical numbers of fluorescent viral complexes per nucleus using the ground truth and predicted nuclear membrane images. We also demonstrate the utility of predicting the nuclear envelope based on transmitted light images for multicolor fluorescence microscopy of infected cells. Importantly, we show that our model can be adapted to predict the nuclear membrane of live cells imaged at 37 °C, making this approach compatible with single virus tracking. Collectively, these findings demonstrate the utility of deep learning approaches for label-free imaging of cellular structures during early stages of virus infection.


Asunto(s)
VIH-1 , Virosis , Humanos , Membrana Nuclear , Transporte Activo de Núcleo Celular , Núcleo Celular , Células HeLa , VIH-1/genética , Replicación Viral/genética
7.
Int J Biol Sci ; 19(12): 3920-3936, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37564209

RESUMEN

Myxofibrosarcoma is genetically complex without established nonsurgical therapies. In public datasets, PAK1 was recurrently gained with mRNA upregulation. Using myxofibrosarcoma cells, we explored the oncogenic underpinning of PAK1 with genetic manipulation and a pan-PAK inhibitor (PF3758309). Myxofibrosarcoma specimens were analyzed for the levels of PAK1, phospho-PAKT423, CSF2 and microvascular density (MVD) and those of PAK1 gene and mRNA. PAK1-expressing xenografts were assessed for the effects of PF3758309 and CSF2 silencing. Besides pro-proliferative and pro-migrator/pro-invasive attributes, PAK1 strongly enhanced angiogenesis in vitro, which, not phenocopied by PAK2-4, was identified as CSF2-mediated using antibody arrays. PAK1 underwent phosphorylation at tyrosines153,201,285 and threonine423 to facilitate nuclear entry, whereby nuclear PAK1 bound STAT5B to co-transactivate the CSF2 promoter, increasing CSF2 secretion needed for angiogenesis. Angiogenesis driven by PAK1-upregulated CSF2 was negated by CSF2 silencing, anti-CSF2, and PF3758309. Clinically, overexpressed whole-cell phospho-PAKT423, related to PAK1 amplification, was associated with increased grades, stages, and PAK1 mRNA, higher MVD, and CSF2 overexpression. Overexpressed whole-cell phospho-PAKT423 and CSF2 independently portended shorter metastasis-free survival and disease-specific survival, respectively. In vivo, both CSF2 silencing and PF3758309 suppressed PAK1-driven tumor proliferation and angiogenesis. Conclusively, the nuclear entry of overexpressed/activated PAK1 endows myxofibrosarcomas with pro-angiogenic function, highlighting the vulnerable PAK1/STAT5B/CSF2 regulatory axis.


Asunto(s)
Fibrosarcoma , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Factor de Transcripción STAT5 , Quinasas p21 Activadas , Humanos , Línea Celular Tumoral , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Fosforilación , ARN Mensajero/metabolismo , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Activación Transcripcional , Animales , Fibrosarcoma/genética , Fibrosarcoma/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
8.
Proc Natl Acad Sci U S A ; 120(13): e2202815120, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36943880

RESUMEN

Increasing evidence has suggested that the HIV-1 capsid enters the nucleus in a largely assembled, intact form. However, not much is known about how the cone-shaped capsid interacts with the nucleoporins (NUPs) in the nuclear pore for crossing the nuclear pore complex. Here, we elucidate how NUP153 binds HIV-1 capsid by engaging the assembled capsid protein (CA) lattice. A bipartite motif containing both canonical and noncanonical interaction modules was identified at the C-terminal tail region of NUP153. The canonical cargo-targeting phenylalanine-glycine (FG) motif engaged the CA hexamer. By contrast, a previously unidentified triple-arginine (RRR) motif in NUP153 targeted HIV-1 capsid at the CA tri-hexamer interface in the capsid. HIV-1 infection studies indicated that both FG- and RRR-motifs were important for the nuclear import of HIV-1 cores. Moreover, the presence of NUP153 stabilized tubular CA assemblies in vitro. Our results provide molecular-level mechanistic evidence that NUP153 contributes to the entry of the intact capsid into the nucleus.


Asunto(s)
Infecciones por VIH , Seropositividad para VIH , VIH-1 , Humanos , Proteínas de la Cápside/metabolismo , Cápside/metabolismo , VIH-1/metabolismo , Transporte Activo de Núcleo Celular , Proteínas de Complejo Poro Nuclear/metabolismo , Infecciones por VIH/metabolismo , Poro Nuclear/metabolismo
9.
J Virol ; 96(17): e0111822, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-35950857

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside , Núcleo Celular , Virus Diminuto del Ratón , Infecciones por Parvoviridae , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Núcleo Celular/virología , Quimotripsina/metabolismo , Ratones , Virus Diminuto del Ratón/fisiología , Infecciones por Parvoviridae/metabolismo , Procesamiento Proteico-Postraduccional
10.
J Cell Physiol ; 237(8): 3181-3204, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35616326

RESUMEN

The PI3K-AKT-MTOR signal transduction pathway is one of the essential signalling cascades within the cell due to its involvement in many vital functions. The pathway initiates with the recruitment of phosphatidylinositol-3 kinases (PI3Ks) onto the plasma membrane, generating phosphatidylinositol-3,4,5-triphosphate [PtdIns(3,4,5)P3 ] and subsequently activating AKT. Being the central node of the PI3K network, AKT activates the mechanistic target of rapamycin kinase complex 1 (MTORC1) via Tuberous sclerosis complex 2 inhibition in the cytoplasm. Although the cytoplasmic role of the pathway has been widely explored for decades, we now know that most of the effector molecules of the PI3K axis diverge from the canonical route and translocate to other cell organelles including the nucleus. The presence of phosphoinositides (PtdIns) inside the nucleus itself indicates the existence of a nuclear PI3K signalling. The nuclear localization of these signaling components is evident in regulating many nuclear processes like DNA replication, transcription, DNA repair, maintenance of genomic integrity, chromatin architecture, and cell cycle control. Here, our review intends to present a comprehensive overview of the nuclear functions of the PI3K-AKT-MTOR signaling biomolecules.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Núcleo Celular , Citoplasma , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol , Proteínas Proto-Oncogénicas c-akt/metabolismo
11.
Methods Mol Biol ; 2483: 339-349, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35286686

RESUMEN

Nuclear entry of cAMP-dependent protein kinase catalytic subunits is typically inferred from changes in net protein amount or kinase activity in the nucleus. Previous methods to directly assess nuclear entry require kinase subunit overexpression and/or supraphysiological cAMP elevation. We describe a method to detect nuclear entry of catalytic subunits expressed at an endogenous level in living cells, stimulated by cAMP in a physiological range, and in real time.


Asunto(s)
Núcleo Celular , Proteínas Quinasas Dependientes de AMP Cíclico , Bioensayo , Dominio Catalítico , Proyectos de Investigación
12.
Front Plant Sci ; 12: 691124, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34630451

RESUMEN

The Salt-Overly-Sensitive (SOS) pathway controls the net uptake of sodium by roots and the xylematic transfer to shoots in vascular plants. SOS3/CBL4 is a core component of the SOS pathway that senses calcium signaling of salinity stress to activate and recruit the protein kinase SOS2/CIPK24 to the plasma membrane to trigger sodium efflux by the Na/H exchanger SOS1/NHX7. However, despite the well-established function of SOS3 at the plasma membrane, SOS3 displays a nucleo-cytoplasmic distribution whose physiological meaning is not understood. Here, we show that the N-terminal part of SOS3 encodes structural information for dual acylation with myristic and palmitic fatty acids, each of which commands a different location and function of SOS3. N-myristoylation at glycine-2 is essential for plasma membrane association and recruiting SOS2 to activate SOS1, whereas S-acylation at cysteine-3 redirects SOS3 toward the nucleus. Moreover, a poly-lysine track in positions 7-11 that is unique to SOS3 among other Arabidopsis CBLs appears to be essential for the correct positioning of the SOS2-SOS3 complex at the plasma membrane for the activation of SOS1. The nuclear-localized SOS3 protein had limited bearing on the salt tolerance of Arabidopsis. These results are evidence of a novel S-acylation dependent nuclear trafficking mechanism that contrasts with alternative subcellular targeting of other CBLs by S-acylation.

13.
Viruses ; 13(8)2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34452291

RESUMEN

HIV-1 can infect non-dividing cells. The nuclear envelope therefore represents a barrier that HIV-1 must traverse in order to gain access to the host cell chromatin for integration. Hence, nuclear entry is a critical step in the early stages of HIV-1 replication. Following membrane fusion, the viral capsid (CA) lattice, which forms the outer face of the retroviral core, makes numerous interactions with cellular proteins that orchestrate the progress of HIV-1 through the replication cycle. The ability of CA to interact with nuclear pore proteins and other host factors around the nuclear pore determines whether nuclear entry occurs. Uncoating, the process by which the CA lattice opens and/or disassembles, is another critical step that must occur prior to integration. Both early and delayed uncoating have detrimental effects on viral infectivity. How uncoating relates to nuclear entry is currently hotly debated. Recent technological advances have led to intense discussions about the timing, location, and requirements for uncoating and have prompted the field to consider alternative uncoating scenarios that presently focus on uncoating at the nuclear pore and within the nuclear compartment. This review describes recent advances in the study of HIV-1 nuclear entry, outlines the interactions of the retroviral CA protein, and discusses the challenges of investigating HIV-1 uncoating.


Asunto(s)
Proteínas de la Cápside/metabolismo , Cápside/metabolismo , Núcleo Celular/virología , Infecciones por VIH/virología , VIH-1/fisiología , Desencapsidación Viral , Animales , Núcleo Celular/metabolismo , VIH-1/genética , Interacciones Huésped-Patógeno , Humanos , Membrana Nuclear/fisiología , Membrana Nuclear/virología , Poro Nuclear/fisiología , Poro Nuclear/virología , Retroviridae/fisiología , Transcripción Reversa
14.
Viruses ; 13(6)2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34204384

RESUMEN

In recent years, major advances in research and experimental approaches have significantly increased our knowledge on the role of the HIV-1 capsid in the virus life cycle, from reverse transcription to integration and gene expression. This makes the capsid protein a good pharmacological target to inhibit HIV-1 replication. This review covers our current understanding of the role of the viral capsid in the HIV-1 life cycle and its interaction with different host factors that enable reverse transcription, trafficking towards the nucleus, nuclear import and integration into host chromosomes. It also describes different promising small molecules, some of them in clinical trials, as potential targets for HIV-1 therapy.


Asunto(s)
Proteínas de la Cápside/genética , Cápside/metabolismo , Infecciones por VIH/virología , VIH-1/fisiología , Integración Viral , Transporte Activo de Núcleo Celular , Proteínas de la Cápside/metabolismo , Núcleo Celular/virología , Interacciones Huésped-Patógeno , Humanos , Replicación Viral
15.
Viruses ; 13(6)2021 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-34203080

RESUMEN

Viruses are pathogens that have evolved to hijack the cellular machinery to replicate themselves and spread to new cells. During the course of evolution, viruses developed different strategies to overcome the cellular defenses and create new progeny. Among them, some RNA and many DNA viruses require access to the nucleus to replicate their genome. In non-dividing cells, viruses can only access the nucleus through the nuclear pore complex (NPC). Therefore, viruses have developed strategies to usurp the nuclear transport machinery and gain access to the nucleus. The majority of these viruses use the capsid to manipulate the nuclear import machinery. However, the particular tactics employed by each virus to reach the host chromatin compartment are very different. Nevertheless, they all require some degree of capsid remodeling. Recent notions on the interplay between the viral capsid and cellular factors shine new light on the quest for the nuclear entry step and for the fate of these viruses. In this review, we describe the main components and function of nuclear transport machinery. Next, we discuss selected examples of RNA and DNA viruses (HBV, HSV, adenovirus, and HIV) that remodel their capsid as part of their strategies to access the nucleus and to replicate.


Asunto(s)
Cápside/metabolismo , Núcleo Celular/metabolismo , Núcleo Celular/virología , Interacciones Microbiota-Huesped , Virus/metabolismo , Transporte Activo de Núcleo Celular , Humanos , Poro Nuclear/virología , Virión/metabolismo , Fenómenos Fisiológicos de los Virus , Replicación Viral
16.
Sci China Life Sci ; 64(1): 66-76, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32430850

RESUMEN

It is recognized that HIV-1 capsid cores are disassembled in the cytoplasm, releasing their genomes into the nucleus through nuclear pores, but there is also evidence showing the capsid (CA) exists in the nucleus. Whether HIV-1 enters the nucleus and how it enters the nucleus through the undersized nuclear pore remains mysterious. Based on multicolor labeling and real-time imaging of the viral and cellular components, our observations via light and electron microscopy suggest that HIV-1 selectively gathered at the microtubule organization center (MTOC), leading the nearby nuclear envelope (NE) to undergo deformation, invagination and restoration to form a nuclear vesicle in which the viral particles were wrapped; then, the inner membrane of the nuclear vesicle ruptured to release HIV-1 into the nucleus. This unexpected discovery expands our understanding of the complexity of HIV-1 nuclear entry, which may provide new insights to HIV-1 virology.


Asunto(s)
Proteínas de la Cápside/metabolismo , Núcleo Celular/metabolismo , Endocitosis , VIH-1/metabolismo , Poro Nuclear/metabolismo , Virión/metabolismo , Transporte Activo de Núcleo Celular , Línea Celular Tumoral , Núcleo Celular/ultraestructura , Núcleo Celular/virología , Células HEK293 , Infecciones por VIH/virología , VIH-1/genética , VIH-1/fisiología , Humanos , Microscopía Confocal , Microscopía Electrónica de Transmisión , Centro Organizador de los Microtúbulos/metabolismo , Centro Organizador de los Microtúbulos/virología , Membrana Nuclear/metabolismo , Membrana Nuclear/ultraestructura , Membrana Nuclear/virología , Poro Nuclear/ultraestructura , Poro Nuclear/virología , Imagen de Lapso de Tiempo/métodos , Virión/ultraestructura
17.
Int J Biochem Cell Biol ; 129: 105875, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33157236

RESUMEN

Viruses are obligatory intracellular parasites. For their efficient replication, many require access to the nuclear interior. Yet, only few viral particles are small enough to passively diffuse through the nuclear pore complexes, calling for alternative strategies to bypass the nuclear envelope barrier. Some viruses will await mitotic nuclear envelope breakdown to gain access, whereas others will exploit more active means, for instance by hijacking nuclear pore transport or by directly targeting constituents of the nuclear envelope so as to remodel and temporarily perturb its integrity. After replication, newly produced viral DNA complexes need to cross the same barrier to exit the nucleus and enter the cytoplasm, where the final stages of virion maturation take place. There are also different flavours to the feat of nuclear egress that vary in delicacy and intensity. In this review, we define the major entry and egress strategies that are exploited by different viruses and describe the molecular details thereof. Ultimately, a deeper understanding of these pathways may help identifying molecular targets for blocking viral reproduction or spreading.


Asunto(s)
Núcleo Celular/virología , Internalización del Virus , Animales , Humanos
18.
Virulence ; 11(1): 1379-1393, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33073687

RESUMEN

Entry of circovirus into the host cell nucleus is essential for viral replication during the early stage of infection. However, the mechanisms by which nucleolar shuttle proteins are used during viral replication is still not well understood. Here, we report a previously unidentified nucleolar localization signal in circovirus capsid protein (Cap), and that circovirus hijacks the nucleolar phosphoprotein nucleophosmin-1 (NPM1) to facilitate its replication. Colocalization analysis showed that NPM1 translocates from the nucleolus to the nucleoplasm and cytoplasm during viral infection. Coimmunoprecipitation and glutathione S-transferase pull-down assays showed that Cap interacts directly with NPM1. Binding domain mapping showed that the arginine-rich N-terminal motif 1MTYPRRRYRRRRHRPRSHLG20 of Cap, and residue serine-48 of the N-terminal oligomerization domain of NPM1, are essential for the interaction. Virus rescue experiments showed that all arginine to alanine substitution in the N-terminal arginine-rich motif of Cap resulted in diminished viral replication. Knockdown of NPM1 and substitution of serine-48 in NPM1 to glutamic acid also decreased viral replication. In addition, binding assays showed that the arginine-rich motif of Cap is a nucleolar localization signal. Taken together, our findings demonstrate that circovirus protein Cap is a nucleolus-located, and regulates viral replication by directly binding to NPM1.


Asunto(s)
Cápside/metabolismo , Circovirus/fisiología , Proteínas Nucleares , Replicación Viral , Animales , Línea Celular , Nucléolo Celular/virología , Células HEK293 , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleofosmina , Unión Proteica , Porcinos
19.
ACS Appl Mater Interfaces ; 12(33): 36882-36894, 2020 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-32666795

RESUMEN

Clinically approved doxorubicin (Dox)-loaded liposomes (e.g., Doxil) guarantee good biosafety, but their insufficient nuclear delivery of Dox (<0.4%) after cellular uptake significantly hampers their final anticancer efficacy. Here, we report that simply doping protoporphyrin IX (PpIX, a commonly used hydrophobic photosensitizer) into the lipid bilayers of Dox-loaded liposomes (the resultant product is termed PpIX/Dox liposomes) is a feasible way to promote the nuclear delivery of Dox. This facile strategy relies on a unique property of PpIX-it presents considerably higher affinity for the real plasma membrane over its liposomal carrier, which drives the doped PpIX molecules to detach from the liposomes when encountering cancer cells. We demonstrate that this process can trigger the efficient release of the loaded Dox molecules and allow them to enter the nuclei of MCF-7 breast cancer cells without being trapped by lysosomes. Regarding the drug-resistant MCF-7/ADR cells, the aberrant activation of the efflux pumps in the plasma membranes expels the internalized Dox. However, we strikingly find that the robust drug resistance can be reversed upon mild laser irradiation because the photodynamic effect of PpIX disrupts the drug efflux system (e.g., P-glycoprotein) and facilitates the nuclear entry of Dox. As a proof-of-concept, this PpIX doping strategy is also applicable for enhancing the effectiveness of cisplatin-loaded liposomes against both A549 and A549/DDP lung cancer cells. In vivo experimental results prove that a single injection of PpIX/Dox liposomes completely impedes the growth of MCF-7 tumors in nude mice within 2 weeks and, in combination with laser irradiation, can synergistically ablate MCF-7/ADR tumors. Biosafety assessments reveal no significant systemic toxicity caused by PpIX/Dox liposomes. This work exemplifies a facile method to modulate the subcellular fate of liposomal drugs and may inspire the optimization of nanopharmaceuticals in the near future.


Asunto(s)
Antineoplásicos/química , Doxorrubicina/análogos & derivados , Liposomas/química , Fármacos Fotosensibilizantes/química , Protoporfirinas/química , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Núcleo Celular/metabolismo , Colesterol/química , Terapia Combinada , Doxorrubicina/química , Doxorrubicina/farmacología , Liberación de Fármacos , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Humanos , Hipertermia Inducida , Liposomas/metabolismo , Ratones Desnudos , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Fotoquimioterapia , Fármacos Fotosensibilizantes/farmacología , Polietilenglicoles/química , Polietilenglicoles/farmacología , Protoporfirinas/farmacología , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Int J Nanomedicine ; 15: 3639-3647, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32547019

RESUMEN

PURPOSE: Astrocyte dysfunction is a hallmark of central nervous system injury or infection. As a primary contributor to neurodegeneration, astrocytes are an ideal therapeutic target to combat neurodegenerative conditions. Gene therapy has arisen as an innovative technique that provides excellent prospect for disease intervention. Poly (lactide-co-glycolide) (PLGA) and polyethylenimine (PEI) are polymeric nanoparticles commonly used in gene delivery, each manifesting their own set of advantages and disadvantages. As a clinically approved polymer by the Federal Drug Administration, well characterized for its biodegradability and biocompatibility, PLGA-based nanoparticles (PLGA-NPs) are appealing for translational gene delivery systems. However, our investigations revealed PLGA-NPs were ineffective at facilitating exogenous gene expression in primary human astrocytes, despite their success in other cell lines. Furthermore, PEI polymers illustrate high delivery efficiency but induce cytotoxicity. The purpose of this study is to develop viable and biocompatible NPsystem for astrocyte-targeted gene therapy. MATERIALS AND METHODS: Successful gene expression by PLGA-NPs alone or in combination with arginine-modified PEI polymers (AnPn) was assessed by a luciferase reporter gene encapsulated in PLGA-NPs. Cytoplasmic release and nuclear localization of DNA were investigated using fluorescent confocal imaging with YOYO-labeled plasmid DNA (pDNA). NP-mediated cytotoxicity was assessed via lactate dehydrogenase in primary human astrocytes and neurons. RESULTS: Confocal imaging of YOYO-labeled pDNA confirmed PLGA-NPs delivered pDNA to the cytoplasm in a dose and time-dependent manner. However, co-staining revealed pDNA delivered by PLGA-NPs did not localize to the nucleus. The addition of AnPn significantly improved nuclear localization of pDNA and successfully achieved gene expression in primary human astrocytes. Moreover, these formulations were biocompatible with both astrocytes and neurons. CONCLUSION: By co-transfecting two polymeric NPs, we developed an improved system for gene delivery and expression in primary human astrocytes. These findings provide a basis for a biocompatible and clinically translatable method to regulate astrocyte function during neurodegenerative diseases and disorders.


Asunto(s)
Arginina/química , Astrocitos/metabolismo , Técnicas de Transferencia de Gen , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , ADN/genética , Células HEK293 , Humanos , Tamaño de la Partícula , Plásmidos/genética , Polietileneimina , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...