Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2666: 55-67, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37166656

RESUMEN

Ribonuclease P (RNase P), which may consist of both protein subunits and a catalytic RNA part, is responsible for 5' maturation of tRNA by cleaving the 5'-leader sequence. In Escherichia coli, RNase P contains a catalytic RNA subunit (M1 RNA) and a protein factor (C5 protein). In human cells, RNase P holoenzyme consists of an RNA subunit (H1 RNA) and multiple protein subunits that include human RPP29 protein. M1GS, a sequence specific targeting ribozyme derived from M1 RNA, can be constructed to target a specific mRNA to degrade it in vitro. Recent studies have shown that M1GS ribozymes are efficient in blocking the expression of viral mRNAs in cultured cells and in animals. These results suggest that RNase P ribozymes have the potential to be useful in basic research and in clinical applications. It has been shown that RNase P binding proteins, such as C5 protein and RPP29, can enhance the activities of M1GS RNA in processing a natural tRNA substrate and a target mRNA. Understanding how RPP29 binds to M1GS RNA and enhances the enzyme's catalytic activity will provide great insight into developing more robust gene-targeting ribozymes for in vivo application. In this chapter, we describe the methods of using Fe(II)-ethylenediaminetetraacetic acid (EDTA) cleavage and nuclease footprint analyses to determine the regions of a M1GS ribozyme that are in proximity to RPP29 protein.


Asunto(s)
ARN Catalítico , Ribonucleasa P , Animales , Humanos , Ribonucleasa P/genética , Ribonucleasa P/metabolismo , ARN Catalítico/metabolismo , Ácido Edético , Subunidades de Proteína/metabolismo , ARN/química , ARN Mensajero/genética , Escherichia coli/metabolismo , Endonucleasas/metabolismo
2.
PLoS Pathog ; 15(2): e1007615, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30779794

RESUMEN

Cytomegalovirus (CMV) infection causes birth defects and life-threatening complications in immunosuppressed patients. Lack of vaccine and need for more effective drugs have driven widespread ongoing therapeutic development efforts against human CMV (HCMV), mostly using murine CMV (MCMV) as the model system for preclinical animal tests. The recent publication (Yu et al., 2017, DOI: 10.1126/science.aam6892) of an atomic model for HCMV capsid with associated tegument protein pp150 has infused impetus for rational design of novel vaccines and drugs, but the absence of high-resolution structural data on MCMV remains a significant knowledge gap in such development efforts. Here, by cryoEM with sub-particle reconstruction method, we have obtained the first atomic structure of MCMV capsid with associated pp150. Surprisingly, the capsid-binding patterns of pp150 differ between HCMV and MCMV despite their highly similar capsid structures. In MCMV, pp150 is absent on triplex Tc and exists as a "Λ"-shaped dimer on other triplexes, leading to only 260 groups of two pp150 subunits per capsid in contrast to 320 groups of three pp150 subunits each in a "Δ"-shaped fortifying configuration. Many more amino acids contribute to pp150-pp150 interactions in MCMV than in HCMV, making MCMV pp150 dimer inflexible thus incompatible to instigate triplex Tc-binding as observed in HCMV. While pp150 is essential in HCMV, our pp150-deletion mutant of MCMV remained viable though with attenuated infectivity and exhibiting defects in retaining viral genome. These results thus invalidate targeting pp150, but lend support to targeting capsid proteins, when using MCMV as a model for HCMV pathogenesis and therapeutic studies.


Asunto(s)
Proteínas de la Cápside/ultraestructura , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiología , Proteínas de la Matriz Viral/metabolismo , Proteínas de la Matriz Viral/fisiología , Animales , Cápside , Proteínas de la Cápside/metabolismo , Microscopía por Crioelectrón/métodos , Citomegalovirus/genética , Citomegalovirus/metabolismo , Citomegalovirus/patogenicidad , Infecciones por Citomegalovirus/metabolismo , Genoma Viral/genética , Humanos , Ratones , Muromegalovirus/metabolismo , Muromegalovirus/patogenicidad , Fosfoproteínas/ultraestructura , Eliminación de Secuencia/genética , Proteínas de la Matriz Viral/ultraestructura , Virión , Ensamble de Virus
3.
RNA ; 25(5): 645-655, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30803999

RESUMEN

External guide sequences (EGSs) signify the short RNAs that induce ribonuclease P (RNase P), an enzyme responsible for processing the 5' termini of tRNA, to specifically cleave a target mRNA by forming a precursor tRNA-like complex. Hence, the EGS technology may serve as a potential strategy for gene-targeting therapy. Our previous studies have revealed that engineered EGS variants induced RNase P to efficiently hydrolyze target mRNAs. In the present research, an EGS variant was designed to be complementary to the mRNA coding for human cytomegalovirus (HCMV) major capsid protein (MCP), which is vital to form the viral capsid. In vitro, the EGS variant was about 80-fold more efficient in inducing human RNase P-mediated cleavage of the target mRNA than a natural tRNA-derived EGS. Moreover, the expressed variant and natural tRNA-originated EGSs led to a decrease of MCP expression by 98% and 73%-74% and a decrease of viral growth by about 10,000- and 200-fold in cells infected with HCMV, respectively. These results reveal direct evidence that the engineered EGS variant has higher efficiency in blocking the expression of HCMV genes and viral growth than the natural tRNA-originated EGS. Therefore, our findings imply that the EGS variant can be a potent candidate agent for the treatment of infections caused by HCMV.


Asunto(s)
Proteínas de la Cápside/genética , Citomegalovirus/genética , ARN Guía de Kinetoplastida/genética , ARN Mensajero/genética , ARN de Transferencia de Serina/genética , ARN Viral/genética , Ribonucleasa P/metabolismo , Emparejamiento Base , Proteínas de la Cápside/biosíntesis , Línea Celular Transformada , Línea Celular Tumoral , Citomegalovirus/metabolismo , Fibroblastos/metabolismo , Fibroblastos/virología , Regulación Viral de la Expresión Génica , Marcación de Gen/métodos , Ingeniería Genética/métodos , Interacciones Huésped-Patógeno/genética , Humanos , Terapia Molecular Dirigida , Neuroglía/metabolismo , Neuroglía/virología , Conformación de Ácido Nucleico , Cultivo Primario de Células , División del ARN , ARN Guía de Kinetoplastida/química , ARN Guía de Kinetoplastida/metabolismo , ARN Mensajero/química , ARN Mensajero/metabolismo , ARN de Transferencia de Serina/química , ARN de Transferencia de Serina/metabolismo , ARN Viral/química , ARN Viral/metabolismo , Ribonucleasa P/química , Ribonucleasa P/genética , Replicación Viral/fisiología
4.
Theranostics ; 8(20): 5634-5644, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30555569

RESUMEN

Rationales: Gene-targeting ribozymes represent promising nucleic acid-based gene interference agents for therapeutic application. We previously used an in vitro selection procedure to engineer novel RNase P-based ribozyme variants with enhanced targeting activity. However, it has not been reported whether these ribozyme variants also exhibit improved activity in blocking gene expression in animals. Methods and Results: In this report, R388-AS, a new engineered ribozyme variant, was designed to target the mRNA of assemblin (AS) of murine cytomegalovirus (MCMV), which is essential for viral progeny production. Variant R338-AS cleaved AS mRNA sequence in vitro at least 200 times more efficiently than ribozyme M1-AS, which originated from the wild type RNase P catalytic RNA sequence. In cultured MCMV-infected cells, R338-AS exhibited better antiviral activity than M1-AS and decreased viral AS expression by 98-99% and virus production by 15,000 fold. In MCMV-infected mice, R388-AS was more active in inhibiting AS expression, blocking viral replication, and improving animal survival than M1-AS. Conclusions: Our results provide the first direct evidence that novel engineered RNase P ribozyme variants with more active catalytic activity in vitro are also more effective in inhibiting viral gene expression in animals. Moreover, our studies imply the potential of engineering novel RNase P ribozyme variants with unique mutations to improve ribozyme activity for therapeutic application.


Asunto(s)
Terapia Genética/métodos , Muromegalovirus/efectos de los fármacos , Muromegalovirus/patogenicidad , ARN Catalítico/genética , Ribonucleasa P/metabolismo , Animales , Citomegalovirus/genética , Ratones , ARN sin Sentido/genética , ARN Mensajero/genética
5.
Viruses ; 10(6)2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29844277

RESUMEN

The CRISPR/Cas9 system has been applied in the genome editing and disruption of latent infections for herpesviruses such as the herpes simplex virus, Epstein⁻Barr virus, cytomegalovirus, and Kaposi's sarcoma-associated herpesvirus. CRISPR/Cas9-directed mutagenesis can introduce similar types of mutations to the viral genome as can bacterial artificial chromosome recombination engineering, which maintains and reconstitutes the viral genome successfully. The cleavage mediated by CRISPR/Cas9 enables the manipulation of disease-associated viral strains with unprecedented efficiency and precision. Additionally, current therapies for herpesvirus productive and latent infections are limited in efficacy and cannot eradicate viruses. CRISPR/Cas9 is potentially adapted for antiviral treatment by specifically targeting viral genomes during latent infections. This review, which focuses on recently published progress, suggests that the CRISPR/Cas9 system is not only a useful tool for basic virology research, but also a promising strategy for the control and prevention of herpesvirus latent infections.


Asunto(s)
Sistemas CRISPR-Cas , Genoma Viral , Infecciones por Herpesviridae/terapia , Herpesviridae/genética , Animales , Citomegalovirus/genética , Edición Génica , Infecciones por Herpesviridae/prevención & control , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/genética , Humanos , Ratones , Mutagénesis , Simplexvirus/genética , Latencia del Virus
6.
Nat Microbiol ; 3(4): 503-513, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29588542

RESUMEN

The precise cell type hosting latent human cytomegalovirus (HCMV) remains elusive. Here, we report that HCMV reprogrammes human haematopoietic progenitor cells (HPCs) into a unique monocyte subset to achieve latency. Unlike conventional monocytes, this monocyte subset possesses higher levels of B7-H4, IL-10 and inducible nitric oxide synthase (iNOS), a longer lifespan and strong immunosuppressive capacity. Cell sorting of peripheral blood from latently infected human donors confirms that only this monocyte subset, representing less than 0.1% of peripheral mononuclear cells, is HCMV genome-positive but immediate-early-negative. Mechanistic studies demonstrate that HCMV promotes the differentiation of HPCs into this monocyte subset by activating cellular signal transducer and activator of transcription 3 (STAT3). In turn, this monocyte subset generates a high level of nitric oxide (NO) to silence HCMV immediate-early transcription and promote viral latency. By contrast, the US28-knockout HCMV mutant, which is incapable of activating STAT3, fails to reprogramme the HPCs and achieve latency. Our findings reveal that via activating the STAT3-iNOS-NO axis, HCMV differentiates human HPCs into a longevous, immunosuppressive monocyte subset for viral latency.


Asunto(s)
Citomegalovirus/inmunología , Citomegalovirus/patogenicidad , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/virología , Interacciones Huésped-Patógeno/inmunología , Tolerancia Inmunológica/genética , Monocitos/virología , Latencia del Virus/inmunología , Diferenciación Celular/fisiología , Reprogramación Celular/genética , Citomegalovirus/genética , Interacciones Huésped-Patógeno/genética , Humanos , Tolerancia Inmunológica/inmunología , Interleucina-10/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Transcripción STAT3/metabolismo , Inhibidor 1 de la Activación de Células T con Dominio V-Set/metabolismo , Latencia del Virus/genética
7.
PLoS Pathog ; 14(1): e1006867, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29377960

RESUMEN

Interferon-γ (IFN-γ) represents one of the most important innate immunity responses in a host to combat infections of many human viruses including human herpesviruses. Human N-myc interactor (Nmi) protein, which has been shown to interact with signal transducer and activator of transcription (STAT) proteins including STAT1, is important for the activation of IFN-γ induced STAT1-dependent transcription of many genes responsible for IFN-γ immune responses. However, no proteins encoded by herpesviruses have been reported to interact with Nmi and inhibit Nmi-mediated activation of IFN-γ immune responses to achieve immune evasion from IFN-γ responses. In this study, we show strong evidence that the UL23 protein of human cytomegalovirus (HCMV), a human herpesvirus, specifically interacts with Nmi. This interaction was identified through a yeast two-hybrid screen and co-immunoprecipitation in human cells. We observed that Nmi, when bound to UL23, was not associated with STAT1, suggesting that UL23 binding of Nmi disrupts the interaction of Nmi with STAT1. In cells overexpressing UL23, we observed (a) significantly reduced levels of Nmi and STAT1 in the nuclei, the sites where these proteins act to induce transcription of IFN-γ stimulated genes, and (b) decreased levels of the induction of the transcription of IFN-γ stimulated genes. UL23-deficient HCMV mutants induced higher transcription of IFN-γ stimulated genes and exhibited lower titers than parental and control revertant viruses expressing functional UL23 in IFN-γ treated cells. Thus, UL23 appears to interact directly with Nmi and inhibit nuclear translocation of Nmi and its associated protein STAT1, leading to a decrease of IFN-γ induced responses and an increase of viral resistance to IFN-γ. Our results further highlight the roles of UL23-Nmi interactions in facilitating viral immune escape from IFN-γ responses and enhancing viral resistance to IFN antiviral effects.


Asunto(s)
Citomegalovirus/fisiología , Evasión Inmune , Inmunidad Innata/efectos de los fármacos , Interferón gamma/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Matriz Viral/fisiología , Células Cultivadas , Citomegalovirus/inmunología , Regulación de la Expresión Génica/inmunología , Células HEK293 , Humanos , Evasión Inmune/efectos de los fármacos , Evasión Inmune/genética , Inmunidad Innata/genética , Unión Proteica , Transducción de Señal/genética , Transducción de Señal/inmunología
8.
Mol Ther Nucleic Acids ; 9: 322-332, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29246310

RESUMEN

External guide sequence (EGS) RNAs are associated with ribonuclease P (RNase P), a tRNA processing enzyme, and represent promising agents for gene-targeting applications as they can direct RNase-P-mediated cleavage of a target mRNA. Using murine cytomegalovirus (MCMV) as a model system, we examined the antiviral effects of an EGS variant, which was engineered using in vitro selection procedures. EGSs were used to target the shared mRNA region of MCMV capsid scaffolding protein (mCSP) and assemblin. In vitro, the EGS variant was 60 times more active in directing RNase P cleavage of the target mRNA than the EGS originating from a natural tRNA. In MCMV-infected cells, the variant reduced mCSP expression by 92% and inhibited viral growth by 8,000-fold. In MCMV-infected mice hydrodynamically transfected with EGS-expressing constructs, the EGS variant was more effective in reducing mCSP expression, decreasing viral production, and enhancing animal survival than the EGS originating from a natural tRNA. These results provide direct evidence that engineered EGS variants with higher targeting activity in vitro are also more effective in reducing gene expression in animals. Furthermore, our findings imply the possibility of engineering potent EGS variants for therapy of viral infections.

9.
PLoS One ; 12(10): e0186791, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29059242

RESUMEN

We have previously engineered new RNase P-based ribozyme variants with improved in vitro catalytic activity. In this study, we employed a novel engineered variant to target a shared mRNA region of human cytomegalovirus (HCMV) immediate early proteins 1 (IE1) and 2 (IE2), which are essential for the expression of viral early and late genes as well as viral growth. Ribozyme F-R228-IE represents a novel variant that possesses three unique base substitution point mutations at the catalytic domain of RNase P catalytic RNA. Compared to F-M1-IE that is the ribozyme derived from the wild type RNase P catalytic RNA sequence, the functional variant F-R228-IE cleaved the target mRNA sequence in vitro at least 100 times more efficiently. In cultured cells, expression of F-R228-IE resulted in IE1/IE2 expression reduction by 98-99% and in HCMV production reduction by 50,000 folds. In contrast, expression of F-M1-IE resulted in IE1/IE2 expression reduction by less than 80% and in viral production reduction by 200 folds. Studies of the ribozyme-mediated antiviral effects in cultured cells suggest that overall viral early and late gene expression and viral growth were inhibited due to the ribozyme-mediated reduction of HCMV IE1 and IE2 expression. Our results provide direct evidence that engineered RNase P ribozymes, such as F-R228-IE, can serve as a novel class of inhibitors for the treatment and prevention of HCMV infection. Moreover, these results suggest that F-R228-IE, with novel and unique mutations at the catalytic domain to enhance ribozyme activity, can be a candidate for the construction of effective agents for anti-HCMV therapy.


Asunto(s)
Citomegalovirus/genética , Genes Inmediatos-Precoces , ARN Catalítico/metabolismo , Ribonucleasa P/metabolismo , Citomegalovirus/crecimiento & desarrollo , Humanos
10.
Sci Rep ; 6: 38451, 2016 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-27941929

RESUMEN

Infection of enterovirus 71 (EV71) and associated hand, foot, and mouth disease (HFMD) are recognized as emerging public health issues worldwide. Hundreds of thousands of children are annually infected with EV71 and develop HFMD in China alone. Studies of EV71 infection are critical to the treatment and prevention of the associated HFMD outbreaks. In this report, we studied an outbreak of 105 HFMD cases in Shawo Township of China between September to October 2012. More than 90% of cases were children younger than 9 years old, with over 50% of cases aged 3-6 years old. Laboratory studies detected a high prevalence of EV71 and suggested EV71 as the most common enterovirus causing HFMD in Shawo. Sequencing analysis showed that the EV71 strains from Shawo belong to the C4 subgenotype, and are phylogenetically more related to those from the distant city of Nanchang than those from the nearby city of Wuhan with distinct variations. More girls were found to be associated with EV71 in Shawo whereas more boys were associated with EV71 in Wuhan and Nanchang. Our studies further the understanding of the molecular epidemiological features of HFMD and infection by enteroviruses in China.


Asunto(s)
Brotes de Enfermedades/estadística & datos numéricos , Enterovirus Humano A/fisiología , Enfermedad de Boca, Mano y Pie/epidemiología , Enfermedad de Boca, Mano y Pie/virología , Distribución por Edad , Secuencia de Bases , Niño , Preescolar , China/epidemiología , Femenino , Geografía , Humanos , Masculino , Filogenia , Estaciones del Año , Alineación de Secuencia , Análisis de Secuencia de ADN
11.
Sci Rep ; 6: 27068, 2016 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-27279482

RESUMEN

An external guide sequence (EGS) is a RNA sequence which can interact with a target mRNA to form a tertiary structure like a pre-tRNA and recruit intracellular ribonuclease P (RNase P), a tRNA processing enzyme, to degrade target mRNA. Previously, an in vitro selection procedure has been used by us to engineer new EGSs that are more robust in inducing human RNase P to cleave their targeted mRNAs. In this study, we constructed EGSs from a variant to target the mRNA encoding herpes simplex virus 1 (HSV-1) major transcription regulator ICP4, which is essential for the expression of viral early and late genes and viral growth. The EGS variant induced human RNase P cleavage of ICP4 mRNA sequence 60 times better than the EGS generated from a natural pre-tRNA. A decrease of about 97% and 75% in the level of ICP4 gene expression and an inhibition of about 7,000- and 500-fold in viral growth were observed in HSV infected cells expressing the variant and the pre-tRNA-derived EGS, respectively. This study shows that engineered EGSs can inhibit HSV-1 gene expression and viral growth. Furthermore, these results demonstrate the potential for engineered EGS RNAs to be developed and used as anti-HSV therapeutics.


Asunto(s)
Herpesvirus Humano 1/genética , Ribonucleasa P/fisiología , Replicación Viral , Expresión Génica , Regulación Viral de la Expresión Génica , Células HeLa , Herpesvirus Humano 1/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Secuencias Invertidas Repetidas , Cinética , Estabilidad del ARN , ARN Guía de Kinetoplastida/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Carga Viral
12.
Viruses ; 7(7): 3345-60, 2015 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-26114473

RESUMEN

An engineered RNase P-based ribozyme variant, which was generated using the in vitro selection procedure, was used to target the overlapping mRNA region of two proteins essential for human cytomegalovirus (HCMV) replication: capsid assembly protein (AP) and protease (PR). In vitro studies showed that the generated variant, V718-A, cleaved the target AP mRNA sequence efficiently and its activity was about 60-fold higher than that of wild type ribozyme M1-A. Furthermore, we observed a reduction of 98%-99% in AP/PR expression and an inhibition of 50,000 fold in viral growth in cells with V718-A, while a 75% reduction in AP/PR expression and a 500-fold inhibition in viral growth was found in cells with M1-A. Examination of the antiviral effects of the generated ribozyme on the HCMV replication cycle suggested that viral DNA encapsidation was inhibited and as a consequence, viral capsid assembly was blocked when the expression of AP and PR was inhibited by the ribozyme. Thus, our study indicates that the generated ribozyme variant is highly effective in inhibiting HCMV gene expression and blocking viral replication, and suggests that engineered RNase P ribozyme can be potentially developed as a promising gene-targeting agent for anti-HCMV therapy.


Asunto(s)
Proteínas de la Cápside/metabolismo , Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , ARN Catalítico/metabolismo , Ribonucleasa P/metabolismo , Replicación Viral , Proteínas de la Cápside/antagonistas & inhibidores , Proteínas de la Cápside/genética , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/terapia , Regulación Viral de la Expresión Génica , Humanos , ARN Catalítico/genética , Ribonucleasa P/genética
13.
Biomed Res Int ; 2013: 509714, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23509733

RESUMEN

External guide sequences (EGSs) represent a new class of RNA-based gene-targeting agents, consist of a sequence complementary to a target mRNA, and render the target RNA susceptible to degradation by ribonuclease P (RNase P). In this study, EGSs were constructed to target the mRNA encoding human CC-chemokine receptor 5 (CCR5), one of the primary coreceptors for HIV. An EGS RNA, C1, efficiently directed human RNase P to cleave the CCR5 mRNA sequence in vitro. A reduction of about 70% in the expression level of both CCR5 mRNA and protein and an inhibition of more than 50-fold in HIV (R5 strain Ba-L) p24 production were observed in cells that expressed C1. In comparison, a reduction of about 10% in the expression of CCR5 and viral growth was found in cells that either did not express the EGS or produced a "disabled" EGS which carried nucleotide mutations that precluded RNase P recognition. Furthermore, the same C1-expressing cells that were protected from R5 strain Ba-L retained susceptibility to X4 strain IIIB, which uses CXCR4 as the coreceptor instead of CCR5, suggesting that the RNase P-mediated cleavage induced by the EGS is specific for the target CCR5 but not the closely related CXCR4. Our results provide direct evidence that EGS RNAs against CCR5 are effective and specific in blocking HIV infection and growth. These results also demonstrate the feasibility to develop highly effective EGSs for anti-HIV therapy.


Asunto(s)
Fármacos Anti-VIH/farmacología , Antagonistas de los Receptores CCR5 , Regulación de la Expresión Génica , VIH-1/fisiología , Ribonucleasa P/metabolismo , Línea Celular , Regulación Viral de la Expresión Génica , Células HeLa , Humanos , Terapia Molecular Dirigida , Mutación , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , ARN Mensajero/antagonistas & inhibidores
14.
Mol Ther ; 21(5): 995-1003, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23481322

RESUMEN

Nucleic acid-based gene interfering approaches, such as those mediated by RNA interference and RNase P-associated external guide sequence (EGS), have emerged as promising antiviral strategies. The RNase P-based technology is unique, because a custom-designed EGS can bind to any complementary mRNA sequence and recruit intracellular RNase P for specific degradation of the target mRNA. In this study, a functional EGS was constructed to target hepatitis B virus (HBV) essential transcripts. Furthermore, an attenuated Salmonella strain was constructed and used for delivery of anti-HBV EGS in cells and in mice. Substantial reduction in the levels of HBV gene expression and viral DNA was detected in cells treated with the Salmonella vector carrying the functional EGS construct. Furthermore, oral inoculation of Salmonella carrying the EGS construct led to an inhibition of ~95% in the levels of HBV gene expression and a reduction of ~200,000-fold in viral DNA level in the livers and sera of the treated mice transfected with a HBV plasmid. Our results suggest that EGSs are effective in inhibiting HBV replication in cultured cells and mammalian livers, and demonstrate the use of Salmonella-mediated delivery of EGS as a promising therapeutic approach for human diseases including HBV infection.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus de la Hepatitis B/fisiología , ARN Pequeño no Traducido/genética , ARN Pequeño no Traducido/metabolismo , Ribonucleasa P/metabolismo , Replicación Viral , Animales , Línea Celular , Expresión Génica , Técnicas de Transferencia de Gen , Genoma Viral , Humanos , Hidrólisis , Hígado/metabolismo , Hígado/patología , Ratones , Conformación de Ácido Nucleico , ARN Pequeño no Traducido/química , ARN Viral/metabolismo , Salmonella/genética , Salmonella/metabolismo , Transfección
15.
RNA Biol ; 9(9): 1186-95, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23018778

RESUMEN

External guide sequences (EGSs) are RNA molecules that can bind to a target mRNA and direct ribonuclease P (RNase P), a tRNA processing enzyme, for specific cleavage of the target mRNA. Using an in vitro selection procedure, we have previously generated EGS variants that efficiently direct human RNase P to cleave a target mRNA in vitro. In this study, we constructed EGSs from a variant to target the overlapping region of the mRNAs coding for human cytomegalovirus (HCMV) capsid scaffolding protein (CSP) and assemblin, which are essential for viral capsid formation. The EGS variant was about 40-fold more active in directing human RNase P to cleave the mRNA in vitro than the EGS derived from a natural tRNA. Moreover, a reduction of about 98% and 75% in CSP/assemblin gene expression and a reduction of 7000- and 250-fold in viral growth were observed in HCMV-infected cells that expressed the variant and the tRNA-derived EGS, respectively. Our study shows that the EGS variant is more effective in blocking HCMV gene expression and growth than the tRNA-derived EGS. Moreover, these results demonstrate the utility of highly active EGS RNA variants in gene targeting applications including anti-HCMV therapy.


Asunto(s)
Citomegalovirus/metabolismo , Regulación Viral de la Expresión Génica , ARN Mensajero/biosíntesis , ARN Viral/biosíntesis , Ribonucleasa P/metabolismo , Línea Celular Tumoral , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/terapia , Terapia Genética/métodos , Humanos , ARN Mensajero/genética , ARN Viral/genética , Ribonucleasa P/genética , ARN Pequeño no Traducido
16.
Proc Natl Acad Sci U S A ; 109(32): 13070-5, 2012 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-22826233

RESUMEN

Ribonuclease P complexed with external guide sequence (EGS) bound to mRNA represents a unique nucleic acid-based gene interference approach for modulation of gene expression. Compared with other strategies, such as RNA interference, the EGS-based technology is unique because a custom-designed EGS molecule can hybridize with any mRNA and recruit intracellular ribonuclease P for specific degradation of the target mRNA. It has not been reported whether the EGS-based technology can modulate gene expression in mice. In this study, a functional EGS was constructed to target the mRNA encoding the protease (mPR) of murine cytomegalovirus (MCMV), which is essential for viral replication. Furthermore, a unique attenuated strain of Salmonella was generated for gene delivery of EGS in cultured cells and in mice. Efficient expression of EGS was observed in cultured cells treated with the generated Salmonella vector carrying constructs with the EGS expression cassette. Moreover, a significant reduction in mPR expression and viral growth was found in MCMV-infected cells treated with Salmonella carrying the construct with the functional EGS sequence. When MCMV-infected mice were orally treated with Salmonella carrying EGS expression cassettes, viral gene expression and growth in various organs of these animals were reduced and animal survival improved. Our study suggests that EGS RNAs, when expressed following Salmonella-mediated gene transfer, effectively inhibit viral gene expression and infection in mice. Furthermore, these results demonstrate the feasibility of developing Salmonella-mediated delivery of EGS as a unique approach for treatment that reduces viral diseases in vivo.


Asunto(s)
Infecciones por Citomegalovirus/prevención & control , Regulación Viral de la Expresión Génica/genética , Muromegalovirus/genética , ARN Mensajero/metabolismo , Ribonucleasa P/metabolismo , Animales , Northern Blotting , Western Blotting , Infecciones por Citomegalovirus/genética , Cartilla de ADN/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Ratones , ARN Mensajero/genética , Ribonucleasa P/genética , Salmonella , ARN Pequeño no Traducido
17.
PLoS One ; 7(12): e51855, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23300569

RESUMEN

Using an in vitro selection procedure, we have previously isolated RNase P ribozyme variants that efficiently cleave an mRNA sequence in vitro. In this study, a variant was used to target the HIV RNA sequence in the tat region. The variant cleaved the tat RNA sequence in vitro about 20 times more efficiently than the wild type ribozyme. Our results provide the first direct evidence that combined mutations at nucleotide 83 and 340 of RNase P catalytic RNA from Escherichia coli (G(83) -> U(83) and G(340) -> A(340)) increase the overall efficiency of the ribozyme in cleaving an HIV RNA sequence. Moreover, the variant is more effective in reducing HIV-1 p24 expression and intracellular viral RNA level in cells than the wild type ribozyme. A reduction of about 90% in viral RNA level and a reduction of 150 fold in viral growth were observed in cells that expressed the variant, while a reduction of less than 10% was observed in cells that either did not express the ribozyme or produced a catalytically inactive ribozyme mutant. Thus, engineered ribozyme variants are effective in inhibiting HIV infection. These results also demonstrate the potential of engineering RNase P ribozymes for anti-HIV application.


Asunto(s)
Regulación Viral de la Expresión Génica , Infecciones por VIH/prevención & control , VIH-1/genética , ARN Catalítico/farmacología , Ribonucleasa P/genética , Replicación Viral/genética , Northern Blotting , Células Cultivadas , Infecciones por VIH/genética , Infecciones por VIH/virología , Humanos , Conformación de Ácido Nucleico , ARN Viral , Ribonucleasa P/metabolismo
18.
Mol Ther ; 19(6): 1116-22, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21427705

RESUMEN

MicroRNAs (miRNAs) are emerging as potential cancer therapeutics, but effective delivery mechanisms to tumor sites are a roadblock to utility. Here we show that systemically delivered, synthetic miRNA mimics in complex with a novel neutral lipid emulsion are preferentially targeted to lung tumors and show therapeutic benefit in mouse models of lung cancer. Therapeutic delivery was demonstrated using mimics of the tumor suppressors, microRNA-34a (miR-34a) and let-7, both of which are often down regulated or lost in lung cancer. Systemic treatment of a Kras-activated autochthonous mouse model of non-small cell lung cancer (NSCLC) led to a significant decrease in tumor burden. Specifically, mice treated with miR-34a displayed a 60% reduction in tumor area compared to mice treated with a miRNA control. Similar results were obtained with the let-7 mimic. These findings provide direct evidence that synthetic miRNA mimics can be systemically delivered to the mammalian lung and support the promise of miRNAs as a future targeted therapy for lung cancer.


Asunto(s)
Emulsiones/química , Vectores Genéticos/química , Lípidos/química , Neoplasias Pulmonares/terapia , MicroARNs/fisiología , Animales , Línea Celular Tumoral , Humanos , Ratones , MicroARNs/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Methods Mol Biol ; 488: 267-77, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18982298

RESUMEN

Ribonuclease P (RNase P) from Escherichia coli is a transfer RNA (tRNA)-processing enzyme and consists of a catalytic RNA subunit (M1 RNA) and a protein component (C5 protein). M1GS, a gene-targeting ribozyme derived from M1 RNA, can cleave a target messenger RNA (mRNA) efficiently in vitro and inhibit its expression effectively in cultured cells. It has been shown that C5 protein can significantly increase the activities of M1 ribozyme and M1GS RNA in cleaving a natural tRNA substrate and a target mRNA, respectively. Understanding how C5 binds to M1GS RNA and affects the specific interactions between the ribozyme and its target mRNA substrates may facilitate the development of gene-targeting ribozymes that function effectively in vivo in the presence of cellular proteins. We describe the methods to determine the regions of a M1GS ribozyme that are potentially in close proximity to C5 protein. Specifically, methods are described in detail in using Fe(II)-ethylenediaminetetraacetic acid (EDTA) cleavage and nuclease footprint analyses to map the regions of the ribozyme in the absence and presence of C5 protein. These methods intend to provide experimental protocols for studying the regions of RNase P ribozyme that are in close contact with C5 protein.


Asunto(s)
Proteínas/química , ARN Catalítico/química , ARN/química , Ribonucleasa P/química , Ácido Edético/química , Compuestos Ferrosos/química , Unión Proteica , Proteínas/metabolismo , ARN/metabolismo , ARN Catalítico/metabolismo , Ribonucleasa P/metabolismo
20.
Proc Natl Acad Sci U S A ; 105(31): 10919-24, 2008 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-18663226

RESUMEN

A functional RNase P ribozyme (M1GS RNA) was constructed to target the overlapping mRNA region of two murine cytomegalovirus (MCMV) capsid proteins essential for viral replication: the assembly protein (mAP) and M80. The customized ribozyme efficiently cleaved the target mRNA sequence in vitro. Moreover, 80% reduction in the expression of mAP and M80 and a 2,000-fold reduction in viral growth were observed in cells expressing the ribozyme. In contrast, there was no significant reduction in viral gene expression and growth in cells that either did not express the ribozyme or produced a "disabled" ribozyme carrying mutations that abolished its catalytic activity. When the ribozyme-expressing constructs were delivered into MCMV-infected SCID mice via a modified "hydrodynamic transfection" procedure, expression of ribozymes was observed in the livers and spleens. Compared with the control animals that did not receive any M1GS constructs or received the disabled ribozyme construct, animals receiving the functional ribozyme construct exhibited a significant reduction of viral gene expression and infection. Viral titers in the spleens, livers, lungs, and salivary glands of the functional ribozyme-treated SCID mice at 21 days after infection were 200- to 2,000-fold lower than those in the control animals. Moreover, survival of the infected animals significantly improved upon receiving the functional ribozyme construct. Our study examines the use of M1GS ribozymes for inhibition of gene expression in animals and demonstrates the utility of RNase P ribozymes for gene targeting applications in vivo.


Asunto(s)
Proteínas de la Cápside/metabolismo , Regulación Viral de la Expresión Génica/genética , Marcación de Gen/métodos , Muromegalovirus/química , ARN Catalítico/metabolismo , Ribonucleasa P/metabolismo , Replicación Viral/genética , Animales , Northern Blotting , Western Blotting , Cartilla de ADN/genética , Ratones , Ratones SCID , Células 3T3 NIH
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...