Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35074918

RESUMEN

MeCP2 is associated with Rett syndrome (RTT), MECP2 duplication syndrome, and a number of conditions with isolated features of these diseases, including autism, intellectual disability, and motor dysfunction. MeCP2 is known to broadly bind methylated DNA, but the precise molecular mechanism driving disease pathogenesis remains to be determined. Using proximity-dependent biotinylation (BioID), we identified a transcription factor 20 (TCF20) complex that interacts with MeCP2 at the chromatin interface. Importantly, RTT-causing mutations in MECP2 disrupt this interaction. TCF20 and MeCP2 are highly coexpressed in neurons and coregulate the expression of key neuronal genes. Reducing Tcf20 partially rescued the behavioral deficits caused by MECP2 overexpression, demonstrating a functional relationship between MeCP2 and TCF20 in MECP2 duplication syndrome pathogenesis. We identified a patient exhibiting RTT-like neurological features with a missense mutation in the PHF14 subunit of the TCF20 complex that abolishes the MeCP2-PHF14-TCF20 interaction. Our data demonstrate the critical role of the MeCP2-TCF20 complex for brain function.


Asunto(s)
Proteína 2 de Unión a Metil-CpG/metabolismo , Complejos Multiproteicos/metabolismo , Trastornos del Neurodesarrollo/etiología , Trastornos del Neurodesarrollo/metabolismo , Factores de Transcripción/metabolismo , Alelos , Animales , Biomarcadores , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Mutación , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica , Sinapsis/metabolismo , Factores de Transcripción/genética
2.
PLoS Pathog ; 18(9): e1010859, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36156097

RESUMEN

The majority of adenovirus (Ad) vectors are based on human Ad type 5, which is a member of Ad species C. Species C also includes the closely-related types 1, 2, 6, 57 and 89. It is known that coagulation factors bind to Ad5 hexon and play a key role in the liver tropism of Ad5 vectors, but it is unclear how coagulation factors affect vectors derived from other species C Ads. We evaluated species C Ad vectors both in vitro and following intravenous injection in mice. To assess the impact of hexon differences, we constructed chimeric Ad5 vectors that contain the hexon hypervariable regions from other species C types, including vectors with hexon mutations that decreased coagulation factor binding. After intravenous injection into mice, vectors with Ad5 or Ad6 hexon had strong liver tropism, while vectors with chimeric hexon from other Ad types had weaker liver tropism due to inhibition by natural antibodies and complement. In addition, we discovered a novel ability of hexon to bind prothrombin, which is the most abundant coagulation factor in blood, and we found striking differences in the affinity of Ads for human, mouse and bovine coagulation factors. When compared to Ad5, vectors with non-Ad5 species C hexons had considerably higher affinity for both human and mouse prothrombin. Most of the vectors tested were strongly dependent on coagulation factors for liver transduction, but vectors with chimeric Ad6 hexon showed much less dependence on coagulation factors than other vectors. We found that in vitro neutralization experiments with mouse serum predicted in vivo behavior of Ad5 vectors, but in vitro experiments did not predict the in vivo behavior of vectors based on other Ad types. In sum, hexons from different human Ad species C viruses confer diverse properties on vectors, including differing abilities to target the liver.


Asunto(s)
Adenovirus Humanos , Protrombina , Adenoviridae , Adenovirus Humanos/genética , Animales , Proteínas de la Cápside/metabolismo , Bovinos , Vectores Genéticos , Humanos , Ratones , Protrombina/genética , Protrombina/metabolismo , Transducción Genética
3.
J Inherit Metab Dis ; 42(6): 1128-1135, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30724386

RESUMEN

The urea cycle and glutamine synthetase (GS) are the two main pathways for waste nitrogen removal and their deficiency results in hyperammonemia. Here, we investigated the efficacy of liver-specific GS overexpression for therapy of hyperammonemia. To achieve hepatic GS overexpression, we generated a helper-dependent adenoviral (HDAd) vector expressing the murine GS under the control of a liver-specific expression cassette (HDAd-GS). Compared to mice injected with a control vector expressing an unrelated reporter gene (HDAd-alpha-fetoprotein), wild-type mice with increased hepatic GS showed reduced blood ammonia levels and a concomitant increase of blood glutamine after intraperitoneal injections of ammonium chloride, whereas blood urea was unaffected. Moreover, injection of HDAd-GS reduced blood ammonia levels at baseline and protected against acute hyperammonemia following ammonia challenge in a mouse model with conditional hepatic deficiency of carbamoyl phosphate synthetase 1 (Cps1), the initial and rate-limiting step of ureagenesis. In summary, we found that upregulation of hepatic GS reduced hyperammonemia in wild-type and Cps1-deficient mice, thus confirming a key role of GS in ammonia detoxification. These results suggest that hepatic GS augmentation therapy has potential for treatment of both primary and secondary forms of hyperammonemia.


Asunto(s)
Amoníaco/metabolismo , Terapia Genética/métodos , Glutamato-Amoníaco Ligasa/genética , Hiperamonemia/genética , Hiperamonemia/terapia , Hígado/metabolismo , Amoníaco/toxicidad , Animales , Carbamoil-Fosfato Sintasa (Amoniaco)/genética , Carbamoil-Fosfato Sintasa (Amoniaco)/metabolismo , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/genética , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/metabolismo , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/terapia , Modelos Animales de Enfermedad , Femenino , Técnicas de Transferencia de Gen , Glutamato-Amoníaco Ligasa/metabolismo , Hiperamonemia/metabolismo , Hiperamonemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos/genética
4.
Mol Genet Metab ; 124(4): 243-253, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29801986

RESUMEN

Carbamoyl phosphate synthetase 1 (CPS1) is a urea cycle enzyme that forms carbamoyl phosphate from bicarbonate, ammonia and ATP. Bi-allelic mutations of the CPS1 gene result in a urea cycle disorder presenting with hyperammonemia, often with reduced citrulline, and without orotic aciduria. CPS1 deficiency is particularly challenging to treat and lack of early recognition typically results in early neonatal death. Therapeutic interventions have limited efficacy and most patients develop long-term neurologic sequelae. Using transgenic techniques, we generated a conditional Cps1 knockout mouse. By loxP/Cre recombinase technology, deletion of the Cps1 locus was achieved in adult transgenic animals using a Cre recombinase-expressing adeno-associated viral vector. Within four weeks from vector injection, all animals developed hyperammonemia without orotic aciduria and died. Minimal CPS1 protein was detectable in livers. To investigate the efficacy of gene therapy for CPS deficiency following knock-down of hepatic endogenous CPS1 expression, we injected these mice with a helper-dependent adenoviral vector (HDAd) expressing the large murine CPS1 cDNA under control of the phosphoenolpyruvate carboxykinase promoter. Liver-directed HDAd-mediated gene therapy resulted in survival, normalization of plasma ammonia and glutamine, and 13% of normal Cps1 expression. A gender difference in survival suggests that female mice may require higher hepatic CPS1 expression. We conclude that this conditional murine model recapitulates the clinical and biochemical phenotype detected in human patients with CPS1 deficiency and will be useful to investigate ammonia-mediated neurotoxicity and for the development of cell- and gene-based therapeutic approaches.


Asunto(s)
Carbamoil-Fosfato Sintasa (Amoniaco)/genética , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/terapia , Terapia Genética , Hiperamonemia/terapia , Amoníaco/metabolismo , Animales , Carbamoil-Fosfato Sintasa (Amoniaco)/uso terapéutico , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/genética , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/metabolismo , Enfermedad por Deficiencia de Carbamoil-Fosfato Sintasa I/patología , Carbamoil Fosfato/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Glutamina/metabolismo , Humanos , Hiperamonemia/genética , Hiperamonemia/metabolismo , Hiperamonemia/patología , Hígado/enzimología , Hígado/patología , Masculino , Ratones , Ratones Noqueados , Mutación , Orotato Fosforribosiltransferasa/deficiencia , Orotato Fosforribosiltransferasa/genética , Orotidina-5'-Fosfato Descarboxilasa/deficiencia , Orotidina-5'-Fosfato Descarboxilasa/genética , Errores Innatos del Metabolismo de la Purina-Pirimidina/genética , Errores Innatos del Metabolismo de la Purina-Pirimidina/patología
5.
Mol Ther ; 21(4): 796-805, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23319058

RESUMEN

We previously dissected the components of the innate immune response to Helper-dependent adenoviral vectors (HDAds) using genetic models, and demonstrated that multiple pattern recognition receptor signaling pathways contribute to this host response to HDAds in vivo. Based on analysis of cytokine expression profiles, type I interferon (IFN) mRNA is induced in host mouse livers at 1 hour post-injection. This type I IFN signaling amplifies cytokine expression in liver independent of the nature of vector DNA sequences after 3 hours post-injection. This type I IFN signaling in response to HDAds administration contributes to transcriptional silencing of both HDAd prokaryotic and eukaryotic DNA in liver. This silencing occurs early and is mediated by epigenetic modification as shown by in vivo chromatin immunoprecipitation (ChIP) with anti-histone deacetylase (HDAC) and promyelocytic leukemia protein (PML). In contrast, self-complementary adeno-associated viral vectors (scAAVs) showed significantly lower induction of type I IFN mRNA in liver compared to HDAds at both early and late time points. These results show that the type I IFN signaling dependent transgene silencing differs between AAV and HDAd vectors after liver-directed gene transfer.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/genética , Interferón Tipo I/genética , Animales , Inmunoprecipitación de Cromatina , Virus Helper/genética , Histona Desacetilasas/metabolismo , Hígado/metabolismo , Ratones , Proteínas Nucleares/metabolismo , Proteína de la Leucemia Promielocítica , Factores de Transcripción/metabolismo , Transgenes/genética , Proteínas Supresoras de Tumor/metabolismo
6.
Mol Ther ; 19(1): 76-82, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20808291

RESUMEN

Murine leukemia virus (MLV)-based replication-competent retrovirus (RCR) vectors have been shown to mediate efficient, selective, and persistent tumor transduction, thereby achieving significant therapeutic benefit in a wide variety of cancer models. To further augment the efficiency of this strategy, we have developed a delivery method employing a gutted adenovirus encoding an RCR vector (AdRCR); thus, tumor cells transduced with the adenoviral vector transiently become RCR vector producer cells in situ. As expected, high-titer AdRCR achieved significantly higher initial transduction levels in human cancer cells both in vitro and in vivo, as compared to the original RCR vector itself. Notably, even at equivalent initial transduction levels, more secondary RCR progeny were produced from AdRCR-transduced cells as compared to RCR-transduced cells, resulting in further acceleration of subsequent RCR replication kinetics. In pre-established tumor models in vivo, prodrug activator gene therapy with high-titer AdRCR could achieve enhanced efficacy compared to RCR alone, in a dose-dependent manner. Thus, AdRCR hybrid vectors offer the advantages of high production titers characteristic of adenovirus and secondary production of RCR in situ, which not only accelerates subsequent vector spread and progressive tumor transduction, but can also significantly enhance the therapeutic efficacy of RCR-mediated prodrug activator gene therapy.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/genética , Virus de la Leucemia Murina/fisiología , Neoplasias/terapia , Neoplasias/virología , Viroterapia Oncolítica/métodos , Retroviridae/fisiología , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Vectores Genéticos/administración & dosificación , Células HeLa , Humanos , Virus de la Leucemia Murina/genética , Ratones , Ratones Desnudos , Neoplasias/genética , Retroviridae/genética , Transducción Genética , Trasplante Heterólogo , Replicación Viral/genética
7.
Mol Ther ; 18(7): 1339-45, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20461064

RESUMEN

Helper-dependent adenoviral vectors (HDAd) are effective tools for liver-directed gene therapy because they can mediate long-term transgene expression in the absence of chronic toxicity. However, high vector doses required for efficient hepatocyte transduction by intravascular delivery result in systemic vector dissemination and dose-dependent activation of the innate immunity. Therefore, strategies to achieve high-efficiency hepatocyte transduction using low vector doses and/or to reduce the acute elevations of proinflammatory cytokines and chemokines may have significant clinical potential. Vasoactive intestinal peptide (VIP) is an endogenous neuropeptide involved in the regulation of hepatic blood flow and plays an important role as modulator of immune functions. Here, we show that VIP pretreatment in mice is able to increase hepatocyte transduction by HDAd, decrease vector uptake by the spleen, reduce elevation of proinflammatory serum cytokines interleukin (IL)-6 and IL-12, and reduce serum levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) following intravenous HDAd injection. VIP pretreatment also resulted in a reduction in the expression of the chemokines macrophage-inflammatory protein 2 (MIP-2), monocyte chemotactic protein 1 (MCP-1), and regulated on activation normal T-cell expressed and secreted (RANTES) in the livers of mice injected with HDAd. These results suggest that VIP can improve the therapeutic index of HDAd by increasing hepatocyte transduction efficiency while reducing cytokine and chemokine expression following intravascular delivery of HDAd.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/administración & dosificación , Hígado/efectos de los fármacos , Hígado/metabolismo , Péptido Intestinal Vasoactivo/farmacología , Animales , Citocinas/metabolismo , Vectores Genéticos/genética , Interleucina-12/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa , Bazo/efectos de los fármacos , Bazo/metabolismo , Transducción Genética
8.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33373331

RESUMEN

Previous studies have shown that nitric oxide (NO) supplements may prevent bone loss and fractures in preclinical models of estrogen deficiency. However, the mechanisms by which NO modulates bone anabolism remain largely unclear. Argininosuccinate lyase (ASL) is the only mammalian enzyme capable of synthesizing arginine, the sole precursor for nitric oxide synthase-dependent (NOS-dependent) NO synthesis. Moreover, ASL is also required for channeling extracellular arginine to NOS for NO production. ASL deficiency (ASLD) is thus a model to study cell-autonomous, NOS-dependent NO deficiency. Here, we report that loss of ASL led to decreased NO production and impairment of osteoblast differentiation. Mechanistically, the bone phenotype was at least in part driven by the loss of NO-mediated activation of the glycolysis pathway in osteoblasts that led to decreased osteoblast differentiation and function. Heterozygous deletion of caveolin 1, a negative regulator of NO synthesis, restored NO production, osteoblast differentiation, glycolysis, and bone mass in a hypomorphic mouse model of ASLD. The translational significance of these preclinical studies was further reiterated by studies conducted in induced pluripotent stem cells from an individual with ASLD. Taken together, our findings suggest that ASLD is a unique genetic model for studying NO-dependent osteoblast function and that the NO/glycolysis pathway may be a new target to modulate bone anabolism.


Asunto(s)
Aciduria Argininosuccínica/metabolismo , Huesos/metabolismo , Diferenciación Celular , Glucólisis , Ácido Nítrico/metabolismo , Osteoblastos/metabolismo , Adolescente , Adulto , Animales , Aciduria Argininosuccínica/genética , Aciduria Argininosuccínica/patología , Huesos/patología , Niño , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Osteoblastos/patología
9.
Mol Ther ; 17(2): 327-33, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19050700

RESUMEN

Helper-dependent adenoviral vectors (HDAd) are devoid of all viral coding sequences and are thus an improvement over early generation Ad because they can provide long-term transgene expression in vivo without chronic toxicity. However, high vector doses are required to achieve efficient hepatic transduction by systemic intravenous injection, and this unfortunately results in dose-dependent acute toxicity. To overcome this important obstacle, we have developed a minimally invasive method to preferentially deliver HDAd into the liver of nonhuman primates. Briefly, a balloon occlusion catheter was percutaneously positioned in the inferior vena cava to occlude hepatic venous outflow. HDAd was injected directly into the occluded liver via a percutaneously placed hepatic artery catheter. Compared to systemic vector injection, this approach resulted in substantially higher hepatic transduction efficiency using clinically relevant low vector doses and was accompanied by mild-to-moderate acute but transient toxicities. Transgene expression was sustained for up to 964 days. These results suggest that our minimally invasive method of delivery can significantly improve the vector's therapeutic index and may be a first step toward clinical application of HDAd for liver-directed gene therapy.


Asunto(s)
Cateterismo/métodos , Terapia Genética/métodos , Vectores Genéticos/genética , Hígado/cirugía , Transducción Genética/métodos , Transgenes/genética , Animales , Vectores Genéticos/toxicidad , Hígado/citología , Hígado/metabolismo , Masculino , Papio
10.
Mol Ther Methods Clin Dev ; 17: 441-447, 2020 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-32154329

RESUMEN

In this study, we developed a single helper-dependent adenovirus (HDAd) to deliver all of the components (donor DNA, CRISPR-associated protein 9 [Cas9], and guide RNA [gRNA]) needed to achieve high-efficiency gene targeting and homology-directed repair in transduced cells. We show that these "all-in-one" HDAds are up to 117-fold more efficient at gene targeting than donor HDAds that do not express CRISPR/Cas9 in human induced pluripotent stem cells (iPSCs). The vast majority (>90%) of targeted recombinants had only one allele targeted, and this was accompanied by high-frequency indel formation in the non-targeted allele at the site of Cas9 cleavage. These indels varied in size and nature, and included large deletions of ∼8 kb. The remaining minority of recombinants had both alleles targeted (so-called bi-allelic targeting). These all-in-one HDAds represent an important platform for accomplishing and expanding the utility of homology-directed repair, especially for difficult-to-transfect cells and for in vivo applications.

11.
Nat Commun ; 11(1): 100, 2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31900387

RESUMEN

Axon initial segments (AISs) generate action potentials and regulate the polarized distribution of proteins, lipids, and organelles in neurons. While the mechanisms of AIS Na+ and K+ channel clustering are understood, the molecular mechanisms that stabilize the AIS and control neuronal polarity remain obscure. Here, we use proximity biotinylation and mass spectrometry to identify the AIS proteome. We target the biotin-ligase BirA* to the AIS by generating fusion proteins of BirA* with NF186, Ndel1, and Trim46; these chimeras map the molecular organization of AIS intracellular membrane, cytosolic, and microtubule compartments. Our experiments reveal a diverse set of biotinylated proteins not previously reported at the AIS. We show many are located at the AIS, interact with known AIS proteins, and their loss disrupts AIS structure and function. Our results provide conceptual insights and a resource for AIS molecular organization, the mechanisms of AIS stability, and polarized trafficking in neurons.


Asunto(s)
Segmento Inicial del Axón/metabolismo , Proteoma/metabolismo , Animales , Axones , Biotinilación , Humanos , Espectrometría de Masas , Ratones , Neuronas/metabolismo , Transporte de Proteínas , Ratas , Ratas Sprague-Dawley
12.
Mol Ther Methods Clin Dev ; 13: 432-439, 2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-31080846

RESUMEN

Prolonged expression of CRISPR/Cas9 raises concerns about off-target cleavage, cytotoxicity, and immune responses. To address these issues, we have developed a system to produce helper-dependent adenoviruses that express CRISPR/Cas9 to direct cleavage of the vectors' own genome after transduction of target cells. To prevent self-cleavage during vector production, it was necessary to downregulate Cas9 mRNA as well as inhibit Cas9 protein activity. Cas9 mRNA downregulation was achieved by inserting the target sequences for the helper-virus-encoded miRNA, mivaRNAI, and producer-cell-encoded miRNAs, hsa-miR183-5p, and hsa-miR218-5p, into the 3' UTR of the HDAd-encoded Cas9 expression cassette. Cas9 protein activity was inhibited by expressing anti-CRISPR proteins AcrIIA2 and AcrAII4 from both the producer cells and the helper virus. After purification, these helper-dependent adenoviruses will perform CRISPR/Cas9-mediated self-cleavage in the transduced target cells, thereby limiting the duration of Cas9 expression and thus represent an important platform for improving the safety of gene editing by CRISPR/Cas9.

13.
Mol Ther Methods Clin Dev ; 15: 285-293, 2019 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-31890728

RESUMEN

We describe a strategy to achieve footprintless bi-allelic homology-directed repair (HDR) using helper-dependent adenoviruses (HDAds). This approach utilizes two HDAds to deliver the donor DNA. These two HDAds are identical except for their selectable marker. One expresses the puromycin N-acetyltransferase-herpes simplex virus I thymidine kinase fusion gene (PACTk), while the other expresses the hygromycin phosphotransferase-herpes simplex virus I thymidine kinase fusion gene (HyTk). Therefore, puromycin and hygromycin double resistance can be used to select for targeted HDAd integration into both alleles. Subsequently, piggyBac-mediated excision of both PACTk and HyTk will confer resistance to gancyclovir, resulting in footprintless HDR at both alleles. However, gene-targeting frequency was not high enough to achieve simultaneous targeting at both alleles. Instead, sequential targeting, whereby the two alleles were targeted one at a time, was required in order to achieve bi-allelic HDR with HDAd.

14.
J Gene Med ; 10(8): 890-6, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18563850

RESUMEN

BACKGROUND: Helper-dependent adenoviral vectors (HDAd) can mediate long-term phenotypic correction in the ornithine transacarbamylase (OTC)-deficient mice model with negligible chronic toxicity. However, the high doses required for metabolic correction will result in systemic inflammatory response syndrome in humans. This acute toxicity represents the major obstacle for clinical applications of HDAd vectors for the treatment of OTC deficiency. Strategies for reducing the dose necessary for disease correction are highly desirable because HDAd acute toxicity is clearly dose-dependent. METHODS: We analysed a potent expression cassette and the hydrodynamic injection for the ability to reduce the HDAd dose necessary for phenotypic correction in OTC-deficient spf-ash mice. RESULTS: We have developed a vector containing a potent expression cassette expressing the OTC transgene, which allowed phenotypic correction at lower doses. Our results suggest that vector expressing greater OTC levels allows correction of orotic acid overproduction at lower doses that make clinical translation more relevant. We were able to further reduce the minimal effective dose by delivering the vector through the hydrodynamic injection technique. CONCLUSIONS: Vectors containing the expression cassette used in the present study, combined with other strategies for improving HDAd therapeutic index, will likely permit application of these vectors for the treatment of OTC deficiency as well as other urea cycle disorders.


Asunto(s)
Adenoviridae/genética , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Terapia Genética/métodos , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa , Ornitina Carbamoiltransferasa/genética , Errores Innatos del Metabolismo de los Aminoácidos/genética , Animales , Modelos Animales de Enfermedad , Vectores Genéticos , Virus Helper/genética , Histocitoquímica , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Mutantes , Ornitina Carbamoiltransferasa/metabolismo , Ácido Orótico/orina , Transgenes
15.
J Assoc Res Otolaryngol ; 9(3): 307-20, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18506528

RESUMEN

Prestin is a membrane protein in the outer hair cell (OHC) that has been shown to be essential for electromotility. OHCs from prestin-null mice do not express prestin, do not have a nonlinear capacitance (the electrical signature of electromotility), and are smaller in size than wild-type OHCs. We sought to determine whether prestin-null OHCs can be transduced to incorporate functional prestin protein in a normal fashion. A recombinant helper-dependent adenovirus expressing prestin and green fluorescent protein (HDAd-prestin-GFP) was created and tested in human embryonic kidney cells (HEK cells). Transduced HEK cells demonstrated membrane expression of prestin and nonlinear capacitance. HDAd-prestin-GFP was then applied to cochlear sensory epithelium explants harvested from wild-type and prestin-null mice at postnatal days 2-3, the age at which native prestin is just beginning to become functional in wild-type mice. At postnatal days 4-5, we investigated transduced OHCs for (1) their prestin expression pattern as revealed by immunofluorescence; (2) their cell surface area as measured by linear capacitance; and (3) their prestin function as indicated by nonlinear capacitance. HDAd-prestin-GFP efficiently transduced OHCs of both genotypes and prestin protein localized to the plasma membrane. Whole-cell voltage clamp studies revealed a nonlinear capacitance in transduced wild-type and prestin-null OHCs, but not in non-transduced cells of either genotype. Prestin transduction did not increase the linear capacitance (cell surface area) for either genotype. In peak nonlinear capacitance, voltage at peak nonlinear capacitance, charge density of the nonlinear capacitance, and shape of the voltage-capacitance curves, the transduced cells of the two genotypes resembled each other and previously reported data from adult wild-type mouse OHCs. Thus, prestin introduced into prestin-deficient OHCs segregates normally to the cell membrane and generates a normal nonlinear capacitance, indicative of normal prestin function.


Asunto(s)
Envejecimiento/metabolismo , Células Ciliadas Auditivas Externas/metabolismo , Proteínas Motoras Moleculares/metabolismo , Órgano Espiral/metabolismo , Adenoviridae/genética , Animales , Animales Recién Nacidos , Línea Celular , Membrana Celular/metabolismo , Electrofisiología , Proteínas Fluorescentes Verdes , Células Ciliadas Auditivas Externas/citología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Motoras Moleculares/genética , Técnicas de Cultivo de Órganos , Órgano Espiral/citología , Técnicas de Placa-Clamp , Transducción Genética
16.
Methods Mol Biol ; 433: 33-53, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18679616

RESUMEN

Helper-dependent adenoviral vectors (HDAd) are deleted of all viral coding sequences and have demonstrated tremendous potential for gene therapy by providing long-term, high-level transgene expression in the absence of chronic toxicity. Thus, HDAd are superior to early generation Ad for gene therapy of genetic diseases where long-term transgene expression is required. This chapter describes in detail the rescue, amplification, and large-scale production of HDAd.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/biosíntesis , Virus Helper/metabolismo , Biología Molecular/métodos , Adenoviridae/aislamiento & purificación , Adenoviridae/fisiología , Recuento de Células , Línea Celular , Vectores Genéticos/aislamiento & purificación , Humanos , Integrasas/metabolismo , Replicación Viral
17.
Methods Mol Biol ; 433: 55-78, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18679617

RESUMEN

Adenoviruses (Ads) possess several features that make them attractive mammalian gene transfer vectors. They can efficiently infect a wide variety of quiescent and proliferating cell types from various species to direct high level viral gene expression, their 36 kb double-stranded DNA genome can be manipulated with relative ease by conventional molecular biology techniques, and they can be readily propagated and purified to yield high titer preparations of very stable virus. Consequently, Ads have been extensively used as vectors for recombinant vaccines, for high-level protein production in cultured cells and for gene therapy where high-level, transient transgene expression is desired. This chapter describes in detail methods for the production and characterization of E1-deleted, first generation adenoviral vectors.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/biosíntesis , Biología Molecular/métodos , Secuencia de Bases , Línea Celular , ADN/genética , Genoma Viral/genética , Humanos , Integrasas/metabolismo , Datos de Secuencia Molecular , Plásmidos/genética , Transcripción Genética , Transfección , Ultracentrifugación
19.
J Phys Chem B ; 111(28): 8119-25, 2007 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-17583934

RESUMEN

Kamlet and Taft polarity parameters are presented for three supercritical solutions containing solutes of different polarities at two concentrations. It is shown that the polarizability and hydrogen-bond-donating ability change significantly with pressure. In the lower pressure regime, naphthalene and salicylic acid cause a decrease in the polarity parameters compared with the pure fluid, whereas toluic acid causes an increase. These differences can be explained in terms of solute-solute and solute-indicator clustering. It is shown that there is, however, a direct correlation between the change in the hydrogen-bond-donating ability and polarizability for all solutes and all pressures and concentrations, which is thought to result from changes in the amount of solvent required to solvate the solute.

20.
J Phys Chem B ; 111(28): 8114-8, 2007 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-17592864

RESUMEN

This study shows that solutes can impart significant structure to supercritical solutions, resulting in unexpectedly high solution viscosity at pressures close to the critical value. The viscosity passes through a minimum as the pressure is increased, and this is accounted for by a solvation of the solutes leading to a decrease in solute-solute interactions. At high pressure, the solution viscosity is similar to that of the pure solvent as solvent-solvent interactions dominate. The increase in relative viscosity is modeled using a modified Dole-Jones equation, and it is shown that the change in relative viscosity is related to the volume fraction occupied by the solute. A general model is presented for simple solutes whereby the viscosity of a supercritical solution can be calculated from the molecular volume of the solute and the viscosity of the pure fluid. The higher than expected viscosity observed at low pressures is used to explain the variation of reaction rate constant with pressure.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA