Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncotarget ; 11(6): 574-588, 2020 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-32110278

RESUMEN

Sepsis is an excessive, dysregulated immune response to infection that activates inflammatory and coagulation cascades, which may lead to tissue injury, multiple organ dysfunction syndrome and death. Millions of individuals die annually of sepsis. To date, the only treatment available is antibiotics, drainage of the infection source when possible, and organ support in intensive care units. Numerous previous attempts to develop therapeutic treatments, directed at discreet targets of the sepsis cascade, could not cope with the complex pathophysiology of sepsis and failed. Here we describe a novel treatment, based on empty capsids of SV40 (nanocapsids - NCs). Studies in a severe rat sepsis model showed that pre-treatment by NCs led to a dramatic increase in survival, from zero to 75%. Transcript analyses (RNAseq) demonstrated that the NC treatment is a paradigm shift. The NCs affect multiple facets of biological functions. The affected genes are modified with time, adjusting to the recovery processes. The NCs effect on normal control rats was negligible. The study shows that the NCs are capable of coping with diseases with intricate pathophysiology. Further studies are needed to determine whether when applied after sepsis onset, the NCs still improve outcome.

2.
Soft Matter ; 16(11): 2803-2814, 2020 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-32104873

RESUMEN

Viruses are remarkable self-assembled nanobiomaterial-based machines, exposed to a wide range of pH values. Extreme pH values can induce dramatic structural changes, critical for the function of the virus nanoparticles, including assembly and genome uncoating. Tuning cargo-capsid interactions is essential for designing virus-based delivery systems. Here we show how pH controls the structure and activity of wild-type simian virus 40 (wtSV40) and the interplay between its cargo and capsid. Using cryo-TEM and solution X-ray scattering, we found that wtSV40 was stable between pH 5.5 and 9, and only slightly swelled with increasing pH. At pH 3, the particles aggregated, while capsid protein pentamers continued to coat the virus cargo but lost their positional correlations. Infectivity was only partly lost after the particles were returned to pH 7. At pH 10 or higher, the particles were unstable, lost their infectivity, and disassembled. Using time-resolved experiments we discovered that disassembly began by swelling of the particles, poking a hole in the capsid through which the genetic cargo escaped, followed by a slight shrinking of the capsids and complete disassembly. These findings provide insight into the fundamental intermolecular forces, essential for SV40 function, and for designing virus-based nanobiomaterials, including delivery systems and antiviral drugs.


Asunto(s)
Proteínas de la Cápside/genética , Genoma Viral/genética , Nanopartículas/química , Virus 40 de los Simios/química , Proteínas de la Cápside/química , Humanos , Concentración de Iones de Hidrógeno , Modelos Moleculares , Nanopartículas/uso terapéutico , Virus 40 de los Simios/genética
3.
ACS Omega ; 4(1): 58-64, 2019 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-30729220

RESUMEN

Multivalent ions affect the structure and organization of virus nanoparticles. Wild-type simian virus 40 (wt SV40) is a nonenveloped virus belonging to the polyomavirus family, whose external diameter is 48.4 nm. Calcium ions and disulfide bonds are involved in the stabilization of its capsid and are playing a role in its assembly and disassembly pathways. Using solution small-angle X-ray scattering (SAXS), we found that the volume of wt SV40 swelled by about 17% when both of its calcium ions were chelated by ethylene glycol-bis(2-aminoethylether)-N,N,N',N'-tetraacetic acid and its disulfide bonds were reduced by dithiothreitol. By applying osmotic stress, the swelling could be reversed. DNA-containing virus-like particles behaved in a similar way. The results provide insight into the structural role of calcium ions and disulfide bonds in holding the capsid proteins in compact conformation.

4.
ACS Nano ; 11(10): 9814-9824, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-28956913

RESUMEN

Crystallization is a fundamental and ubiquitous process that is well understood in the case of atoms or small molecules, but its outcome is still hard to predict in the case of nanoparticles or macromolecular complexes. Controlling the organization of virus nanoparticles into a variety of 3D supramolecular architectures is often done by multivalent ions and is of great interest for biomedical applications such as drug or gene delivery and biosensing, as well as for bionanomaterials and catalysis. In this paper, we show that slow dialysis, over several hours, of wild-type Simian Virus 40 (wt SV40) nanoparticle solution against salt solutions containing MgCl2, with or without added NaCl, results in wt SV40 nanoparticles arranged in a body cubic center crystal structure with Im3m space group, as a thermodynamic product, in coexistence with soluble wt SV40 nanoparticles. The nanoparticle crystals formed above a critical MgCl2 concentrations. Reentrant melting and resolubilization of the virus nanoparticles took place when the MgCl2 concentrations passed a second threshold. Using synchrotron solution X-ray scattering we determined the structures and the mass fraction of the soluble and crystal phases as a function of MgCl2 and NaCl concentrations. A thermodynamic model, which balances the chemical potentials of the Mg2+ ions in each of the possible states, explains our observations. The model reveals the mechanism of both the crystallization and the reentrant melting and resolubilization and shows that counterion entropy is the main driving force for both processes.


Asunto(s)
Nanopartículas/química , Virus 40 de los Simios/química , Termodinámica , Cristalización , Virus 40 de los Simios/aislamiento & purificación , Solubilidad
5.
Oncotarget ; 7(33): 52643-52660, 2016 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-27462916

RESUMEN

SV40 large T-antigen (T-ag) has been known for decades to inactivate the tumor suppressor p53 by sequestration and additional mechanisms. Our present study revealed that the struggle between p53 and T-ag begins very early in the infection cycle. We found that p53 is activated early after SV40 infection and defends the host against the infection. Using live cell imaging and single cell analyses we found that p53 dynamics are variable among individual cells, with only a subset of cells activating p53 immediately after SV40 infection. This cell-to-cell variabilty had clear consequences on the outcome of the infection. None of the cells with elevated p53 at the beginning of the infection proceeded to express T-ag, suggesting a p53-dependent decision between abortive and productive infection. In addition, we show that artificial elevation of p53 levels prior to the infection reduces infection efficiency, supporting a role for p53 in defending against SV40. We further found that the p53-mediated host defense mechanism against SV40 is not facilitated by apoptosis nor via interferon-stimulated genes. Instead p53 binds to the viral DNA at the T-ag promoter region, prevents its transcriptional activation by Sp1, and halts the progress of the infection. These findings shed new light on the long studied struggle between SV40 T-ag and p53, as developed during virus-host coevolution. Our studies indicate that the fate of SV40 infection is determined as soon as the viral DNA enters the nucleus, before the onset of viral gene expression.


Asunto(s)
Antígenos Transformadores de Poliomavirus/genética , Regulación Viral de la Expresión Génica , Virus 40 de los Simios/genética , Proteína p53 Supresora de Tumor/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/virología , Línea Celular , Regulación Neoplásica de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Células MCF-7 , Microscopía Confocal , Regiones Promotoras Genéticas/genética , Unión Proteica , Virus 40 de los Simios/fisiología , Factor de Transcripción Sp1/metabolismo , Imagen de Lapso de Tiempo/métodos , Proteína p53 Supresora de Tumor/metabolismo
6.
Oncotarget ; 7(13): 15369-81, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26992229

RESUMEN

Polyomaviruses are a diverse family of viruses which are prevalent in the human population. However, the interactions of these viruses with the immune system are not well characterized. We have previously shown that two human polyomaviruses, JC and BK, use an identical microRNA to evade immune attack by Natural Killer (NK) cells. We showed that this viral microRNA suppresses ULBP3 expression, a stress induced ligand for the killer receptor NKG2D. Here we show that Simian Virus 40 (SV40) also evades NK cell attack through the down regulation of another stress-induced ligand of NKG2D, ULBP1. These findings indicate that NK cells play an essential role in fighting polyomavirus infections and further emphasize the importance of various members of the ULBP family in controlling polyomavirus infection.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Evasión Inmune/inmunología , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Células Asesinas Naturales/inmunología , Infecciones por Polyomavirus/inmunología , Infecciones Tumorales por Virus/inmunología , Animales , Línea Celular , Regulación hacia Abajo , Proteínas Ligadas a GPI/biosíntesis , Humanos , Virus 40 de los Simios/inmunología
7.
Cell Host Microbe ; 14(1): 63-73, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23870314

RESUMEN

A pathogen's ability to engage host receptors is a critical determinant of its host range and interspecies transmissibility, key issues for understanding emerging diseases. However, the identification of host receptors, which are also attractive drug targets, remains a major challenge. Our structural bioinformatics studies reveal that both bacterial and viral pathogens have evolved to structurally mimic native host ligands (ligand mimicry), thus enabling engagement of their cognate host receptors. In contrast to the structural homology, amino acid sequence similarity between pathogen molecules and the mimicked host ligands was low. We illustrate the utility of this concept to identify pathogen receptors by delineating receptor tyrosine kinase Axl as a candidate receptor for the polyomavirus SV40. The SV40-Axl interaction was validated, and its participation in the infection process was verified. Our results suggest that ligand mimicry is widespread, and we present a quick tool to screen for pathogen-host receptor interactions.


Asunto(s)
Bacterias/metabolismo , Infecciones Bacterianas/metabolismo , Receptores de Superficie Celular/química , Receptores Virales/química , Virosis/metabolismo , Virus/metabolismo , Algoritmos , Animales , Bacterias/genética , Infecciones Bacterianas/genética , Infecciones Bacterianas/microbiología , Interacciones Huésped-Patógeno , Humanos , Ligandos , Modelos Moleculares , Unión Proteica , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo , Homología de Secuencia de Aminoácido , Virosis/genética , Virosis/virología , Virus/genética
8.
Nucleus ; 2(4): 320-30, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21941111

RESUMEN

The canonical gate of viruses and viral genomes into the nucleus in non-dividing cells is the nuclear pore, embedded within the nuclear envelope. However, we found that for SV40, the nuclear envelope poses a major hurdle to infection: FISH analysis revealed that the majority of viral DNA remains trapped in the ER; silencing of Lamin A/C rendered the cells more susceptible to infection; and proliferating cells are more susceptible to infection than quiescent cells. Surprisingly, we observed that following SV40 infection the nuclear envelope, including lamins A/C, B1, B2 and the nuclear pore complex, was dramatically deformed, as seen by immunohistochemistry. The infection induced fluctuations in the level of lamin A/C, dephosphorylation of an unknown epitope and leakage to the cytoplasm just prior to and during nuclear entry. Deformations were transient, and the spherical structure of the nuclear envelope was restored subsequent to nuclear entry. Nuclear envelope deformations and lamin A/C dephosphorylation depended on caspase-6 cleavage of lamin A/C. Notably, we have previously reported that inhibition of caspase-6 abolishes SV40 infection. Taken together the results suggest that alterations of the nuclear lamina, induced by the infecting virus, are involved in the nuclear entry of the SV40 genome. We propose that SV40 utilize this unique, previously unknown mechanism for direct trafficking of its genome from the ER to the nucleus. As SV40 serves as a paradigm for the pathogenic human BK, JC and Merkel cell polyomavirus, this study suggests nuclear entry as a novel drug target for these infections.


Asunto(s)
Lamina Tipo A/metabolismo , Membrana Nuclear/fisiología , Virus 40 de los Simios/metabolismo , Animales , Caspasa 6/metabolismo , Línea Celular , Chlorocebus aethiops , Genoma Viral/fisiología , Células HEK293 , Humanos , Inmunohistoquímica , Lamina Tipo A/genética , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Poro Nuclear/metabolismo , Virus 40 de los Simios/genética , Internalización del Virus
9.
J Virol Methods ; 164(1-2): 145-7, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20005903

RESUMEN

SV40 titer is determined traditionally by the conventional plaque assay. Plaques appear after several rounds of infection and the assay takes around two weeks, which may delay research. A simpler assay was developed, based on detection of T-antigen in the infected cells by flow cytometry. Cells grown in 6-well plates are infected with serial dilutions of the viral stock, harvested 48h post-infection, stained and analyzed for T-antigen using a flow cytometer. The viral titer is calculated based on the percentage of T-antigen positive cells. The procedure is accomplished in 2 days. Unexpectedly we found that titers on different permissive African Green Monkey kidney cell lines were consistently different, suggesting variable susceptibility to SV40 infection. The method described, optimized for SV40 titration, may be adapted readily to other viruses.


Asunto(s)
Citometría de Flujo/métodos , Virus 40 de los Simios/aislamiento & purificación , Carga Viral/métodos , Animales , Antígenos Transformadores de Poliomavirus/análisis , Línea Celular , Chlorocebus aethiops , Factores de Tiempo
10.
J Mol Biol ; 386(5): 1382-91, 2009 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-19094992

RESUMEN

The simian virus 40 (SV40) outer shell is composed of 72 pentamers of VP1. The core of the VP1 monomer is a beta-barrel with jelly-roll topology and extending N- and C-terminal arms. A pentapeptide hinge, KNPYP, tethers the C-arm to the VP1 beta-barrel core. The five C-arms that extend from each pentamer insert into the neighbouring pentamers, tying them together through different types of interactions. In the mature virion, this element adopts either of six conformations according to their location in the capsid. We found that the hinge is conserved among 16 members of the Polyomaviridae, attesting to its importance in capsid assembly and/or structure. We have used site-directed mutagenesis to gain an understanding into the structural requirements of this element: Y299 was changed to A, F, and T, and P300 to A and G. The mutants showed reduction in viability to varying degrees. Unexpectedly, assembly was reduced only to a small extent. However, the data showed that the mutants were highly unstable. The largest effect was observed for mutations of P300, indicating a role of the proline in the virion structure. P300G was more unstable than P300A, indicating a requirement for rigidity of the pentapeptide hinge. Y299T and Y299A were more defective in viability than Y299F, highlighting the importance of an aromatic ring at this position. Structural inspection showed that this aromatic ring contacts C-arms of neighbouring pentamers. Computational modelling predicted loss of stability of the Y mutants in concordance with the experimental results. This study provides insights into the structural details of the pentapeptide hinge that are responsible for capsid stability.


Asunto(s)
Proteínas de la Cápside/fisiología , Cápside/fisiología , Modelos Moleculares , Oligopéptidos/fisiología , Virus 40 de los Simios/fisiología , Secuencia de Aminoácidos , Animales , Proteínas de la Cápside/genética , Línea Celular , Secuencia Conservada , Viabilidad Microbiana , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Conformación Proteica , Virión/fisiología , Ensamble de Virus , Replicación Viral
11.
Pancreas ; 36(4): 411-6, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18437088

RESUMEN

OBJECTIVES: Viral vector uptake into the pancreas is rare. The few viral vectors reported to transduce in vivo pancreatic islets after systemic injection required additional physical measures, such as direct pancreatic injection or hepatic vessel clamping. Because pancreatic islet uptake of the human polyomavirus family member BK virus was previously reported in hamsters after systemic administration, we hypothesized that SV40, a polyomavirus member remarkably similar to BK virus, may also infect the pancreas. METHODS: We injected intravenously a low dose of SV40, unaided by any other physical or chemical means, and evaluated viral uptake by pancreatic islets and pancreatic exocrine tissue via polymerase chain reaction, Western blot, electron microscopy, immunofluorescent microscopy, and protein A-gold immunocytochemistry. RESULTS: Pancreatic uptake of SV40 was comparable to other major organs (ie, liver and spleen). SV40 viral particles were detected in both pancreatic islets and acini. In pancreatic islets, all islet cell types were infected by SV40, albeit the infection rate of glucagon-producing alpha cells surpassed beta- and delta-islet cells. Low-dose SV40 administration was not sufficient to induce heterologous gene expression in the pancreas. CONCLUSIONS: Our study shows that pancreatic islet and acinar cell uptake of SV40 is feasible with a single, low-dose intravenous injection. However, this dose did not result in gene delivery into the murine pancreas.


Asunto(s)
Islotes Pancreáticos/virología , Enfermedades Pancreáticas/virología , Virus 40 de los Simios/patogenicidad , Animales , Diabetes Mellitus Tipo 1/virología , Femenino , Regulación de la Expresión Génica , Islotes Pancreáticos/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Enfermedades Pancreáticas/patología , Reacción en Cadena de la Polimerasa , Virus 40 de los Simios/aislamiento & purificación , Virus 40 de los Simios/ultraestructura
12.
PLoS One ; 2(8): e765, 2007 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-17712413

RESUMEN

SV40 is a small, non enveloped DNA virus with an icosahedral capsid of 45 nm. The outer shell is composed of pentamers of the major capsid protein, VP1, linked via their flexible carboxy-terminal arms. Its morphogenesis occurs by assembly of capsomers around the viral minichromosome. However the steps leading to the formation of mature virus are poorly understood. Intermediates of the assembly reaction could not be isolated from cells infected with wt SV40. Here we have used recombinant VP1 produced in insect cells for in vitro assembly studies around supercoiled heterologous plasmid DNA carrying a reporter gene. This strategy yields infective nanoparticles, affording a simple quantitative transduction assay. We show that VP1 assembles under physiological conditions into uniform nanoparticles of the same shape, size and CsCl density as the wild type virus. The stoichiometry is one DNA molecule per capsid. VP1 deleted in the C-arm, which is unable to assemble but can bind DNA, was inactive indicating genuine assembly rather than non-specific DNA-binding. The reaction requires host enzymatic activities, consistent with the participation of chaperones, as recently shown. Our results demonstrate dramatic cooperativity of VP1, with a Hill coefficient of approximately 6. These findings suggest that assembly may be a concerted reaction. We propose that concerted assembly is facilitated by simultaneous binding of multiple capsomers to a single DNA molecule, as we have recently reported, thus increasing their local concentration. Emerging principles of SV40 assembly may help understanding assembly of other complex systems. In addition, the SV40-based nanoparticles described here are potential gene therapy vectors that combine efficient gene delivery with safety and flexibility.


Asunto(s)
Proteínas de la Cápside/metabolismo , Virus 40 de los Simios/metabolismo , Ensamble de Virus , Animales , Cápside/química , Cápside/metabolismo , Cápside/ultraestructura , Proteínas de la Cápside/genética , Proteínas de la Cápside/ultraestructura , ADN Viral/genética , ADN Viral/metabolismo , Terapia Genética , Vectores Genéticos , Humanos , Nanopartículas , Virus 40 de los Simios/química , Virus 40 de los Simios/genética
13.
J Hepatol ; 40(3): 520-6, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15123369

RESUMEN

BACKGROUND/AIMS: Chronic HBV infection, a world-wide epidemic, can lead to chronic hepatitis and eventually to cirrhosis and hepatocellular carcinoma. The liver poses obstacles for many available gene-transfer vectors. SV40-based vectors can transduce human hepatic and hematopoietic cells. We studied the effect of HBV on the transduction - efficiency of human hepatic cells by SV40 - based vectors. METHODS: A SV40-vector carrying the luciferase gene, and wild-type SV40, were used to assess transduction efficiency of human HBV-positive and HBV-negative hepatic cells. Transduction efficiency was measured as luciferase activity or by T-antigen staining. To evaluate whether differences in transduction efficiency are due to cell recognition and/or nuclear transport, MHC-I receptors were measured by FACS analysis and SV40-DNA was extracted from the nuclei of transduced cells and quantified. RESULTS: Two HBV-positive cell-lines, HepG2.2.2.15 and FLC4-A10II, were transduced significantly more efficiently than their parental HBV-negative cell-lines. Transient transfection of HuH-7 cells with the HBV genome also increased transduction efficiency. The level of MHC-I, the cellular receptor for SV40, was comparable in all the cell-lines studied. However, soon after infection with SV40, the nuclei of HepG2.2.2.15 contained >6-fold more SV40-DNA than HepG2. CONCLUSIONS: HBV increases transduction by SV40-vectors. This is due to enhanced vector entry and/or transport into the nucleus. SV40-vectors appear to have a potential for gene therapy for the treatment of HBV infections.


Asunto(s)
Vectores Genéticos , Virus de la Hepatitis B/fisiología , Hepatocitos/fisiología , Virus 40 de los Simios/genética , Transducción Genética , Línea Celular , Núcleo Celular/metabolismo , ADN Viral/metabolismo , Dimerización , Expresión Génica , Genoma Viral , Virus de la Hepatitis B/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Infecciones por Polyomavirus/metabolismo , Regiones Promotoras Genéticas , Receptores Virales/metabolismo , Transfección , Infecciones Tumorales por Virus/metabolismo , Proteínas Virales/metabolismo
14.
Virology ; 304(2): 155-9, 2002 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-12504557

RESUMEN

Simian virus 40 (SV40) vectors are efficient vehicles for gene delivery to hematopoietic and hepatic cells. To ensure their replication incompetence and because of safety considerations, it is critical that the vectors do not contain T-antigen sequences. Available packaging cell lines for T-antigen replacement vectors, COS and CMT4, contain considerable sequence identity with the vectors, leading to homologous recombination and reacquisition of the T-antigen gene. We constructed a packaging cell line, COT18, with minimal sequence identity to the vector. Vector stocks produced by passaging on COT18 had high transducing activity and undetectable levels of T-antigen-positive, replication-competent contaminants. This cell line provides a means for the preparation of safe SV40 vector stocks.


Asunto(s)
Antígenos Transformadores de Poliomavirus/fisiología , Vectores Genéticos/genética , Recombinación Genética , Virus 40 de los Simios/genética , Replicación Viral , Animales , Células COS , Chlorocebus aethiops , Virus 40 de los Simios/fisiología , Transducción Genética , Células Vero
15.
J Virol ; 76(12): 5915-24, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12021324

RESUMEN

Simian virus 40 (SV40) capsid assembly occurs in the nucleus. All three capsid proteins bind DNA nonspecifically, raising the dilemma of how they attain specificity to the SV40 minichromosome in the presence of a large excess of genomic DNA. The SV40 packaging signal, ses, which is required for assembly, is composed of multiple DNA elements that bind transcription factor Sp1. Our previous studies showed that Sp1 participates in SV40 assembly and that it cooperates in DNA binding with VP2/3. We hypothesized that Sp1 recruits the capsid proteins to the viral minichromosome, conferring upon them specific DNA recognition. Here, we have tested the hypothesis. Computer analysis showed that the combination of six tandem GC boxes at ses is not found at cellular promoters and therefore is unique to SV40. Cooperativity in DNA binding between Sp1 and VP2/3 was not abolished at even a 1,000-fold excess of cellular DNA, providing strong support for the recruitment hypothesis. Sp1 also binds VP1 and cooperates with VP1 in DNA binding. VP1 pentamers (VP1(5)) avidly interact with VP2/3, utilizing the same VP2/3 domain as described for polyomavirus. We conclude that VP1(5)-VP2/3 building blocks are recruited by Sp1 to ses, where they form the nucleation center for capsid assembly. By this mechanism the virus ensures that capsid formation is initiated at a single site around its minichromosome. Sp1 enhances the formation of SV40 pseudovirions in vitro, providing additional support for the model. Analyses of Sp1 and VP3 deletion mutants showed that Sp1 and VP2/3 bind one another and cooperate in DNA binding through their DNA-binding domains, with additional contacts outside these domains. VP1 contacts Sp1 at residues outside the Sp1 DNA-binding domain. These and additional data allowed us to propose a molecular model for the VP1(5)-VP2/3-DNA-Sp1 complex.


Asunto(s)
Proteínas de la Cápside , Cápside/metabolismo , Elementos Transponibles de ADN/genética , Regulación Viral de la Expresión Génica , Factor de Transcripción Sp1/metabolismo , Ensamble de Virus/genética , Sitios de Unión , ADN Viral/genética , ADN Viral/metabolismo , Células HeLa , Humanos , Secuencias Reguladoras de Ácidos Nucleicos , Virus 40 de los Simios/metabolismo , Factor de Transcripción Sp1/genética
16.
Hum Gene Ther ; 13(2): 299-310, 2002 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-11812285

RESUMEN

A procedure for in vitro packaging of plasmid DNA in recombinant SV40 capsid proteins was developed by Sandalon et al. (1997). Here, we report the highly efficient transduction into different human, murine and monkey cell lines using a scaled-up protocol for producing SV40 pseudovirions, packaged in vitro, carrying the human multidrug-resistance gene MDR1 encoding P-glycoprotein (P-gp) or the green fluorescent protein reporter gene (GFP) under control of SV40 and cytomegalovirus (CMV) promoters. The percentage of expressing cells was proportional to the number of transducing particles, with close to 100% of cells transduced at optimal ratios of transducing particles to cells. The ability to confer multidrug resistance was evaluated by measuring dye efflux and cell-surface expression in infected cells. The relative level of expression of P-gp driven by the different promoters varied among different cell lines. In human lymphoblastoid cells, which express high levels of major histocompatibility complex (MHC) class I (a surface receptor for SV40), constructs that carry an intron yield the highest expression. Our experiments further demonstrate that MDR1 and GFP expression driven by these promoters is transient; however, transduced cells remain MDR1-positive if selected in colchicine. Thus, the SV40 vectors are well suited to situations in which only short-term expression is required or expression is selected, such as for bone marrow protection during chemotherapy.


Asunto(s)
Colchicina/análogos & derivados , Vectores Genéticos , Virus 40 de los Simios/genética , Transducción Genética , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Colchicina/farmacología , Genes MDR/genética , Proteínas Fluorescentes Verdes , Células HeLa , Antígenos de Histocompatibilidad Clase I/biosíntesis , Humanos , Proteínas Luminiscentes/genética , Ratones , Virus 40 de los Simios/efectos de los fármacos , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...