Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
Más filtros

Banco de datos
País/Región como asunto
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 Intensive Care Med ; 36(3): 373-375, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32935610

RESUMEN

BACKGROUND: In the setting of the COVID pandemic, many patients falling ill with acute respiratory distress syndrome eventually require prone positioning for gas exchange. Traditionally, central venous catheters are inserted with patient in the supine or Trendelenburg position. However, when a patient cannot tolerate supine position and the need for central venous access is urgent, catheter placement may be considered with the patient in the prone position. CASE SUMMARY: A 69-year-old male with rapidly declining respiratory status secondary to COVID pneumonia quickly developed acute respiratory distress syndrome, was rapidly intubated, and then placed in the prone position. Patient could not tolerate the supine position even briefly and required a central venous catheter insertion for continuous renal replacement therapy. We kept the patient in the prone position and successfully inserted a central venous catheter in such position with real-time ultrasound guidance and using micropuncture technique. CONCLUSION: In the setting of the COVID pandemic, many cases of acute respiratory distress syndrome require patients to be prone in order to improve gas exchange. In the most severe situations, these patients would not be able to tolerate rotating back to the supine position but would still require central venous catheter insertion urgently. We demonstrated feasibility of central venous catheter insertion in the prone position in these severely ill patients.


Asunto(s)
COVID-19/terapia , Cateterismo Venoso Central/métodos , Posicionamiento del Paciente/métodos , Posición Prona , Síndrome de Dificultad Respiratoria/terapia , Ultrasonografía Intervencional/métodos , Anciano , Humanos , Intubación Intratraqueal , Masculino , Punciones , SARS-CoV-2
4.
Genome Res ; 27(3): 335-348, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27965292

RESUMEN

Half the human genome is made of transposable elements (TEs), whose ongoing activity continues to impact our genome. LINE-1 (or L1) is an autonomous non-LTR retrotransposon in the human genome, comprising 17% of its genomic mass and containing an average of 80-100 active L1s per average genome that provide a source of inter-individual variation. New LINE-1 insertions are thought to accumulate mostly during human embryogenesis. Surprisingly, the activity of L1s can further impact the somatic human brain genome. However, it is currently unknown whether L1 can retrotranspose in other somatic healthy tissues or if L1 mobilization is restricted to neuronal precursor cells (NPCs) in the human brain. Here, we took advantage of an engineered L1 retrotransposition assay to analyze L1 mobilization rates in human mesenchymal (MSCs) and hematopoietic (HSCs) somatic stem cells. Notably, we have observed that L1 expression and engineered retrotransposition is much lower in both MSCs and HSCs when compared to NPCs. Remarkably, we have further demonstrated for the first time that engineered L1s can retrotranspose efficiently in mature nondividing neuronal cells. Thus, these findings suggest that the degree of somatic mosaicism and the impact of L1 retrotransposition in the human brain is likely much higher than previously thought.


Asunto(s)
Elementos Transponibles de ADN , Elementos de Nucleótido Esparcido Largo , Células-Madre Neurales/metabolismo , División Celular , Células Cultivadas , Células HeLa , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Mosaicismo , Células-Madre Neurales/citología
5.
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
6.
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
7.
Blood ; 128(18): 2206-2217, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27554082

RESUMEN

Current protocols for hematopoietic stem/progenitor cell (HSPC) gene therapy, involving the transplantation of ex vivo genetically modified HSPCs are complex and not without risk for the patient. We developed a new approach for in vivo HSPC transduction that does not require myeloablation and transplantation. It involves subcutaneous injections of granulocyte-colony-stimulating factor/AMD3100 to mobilize HSPCs from the bone marrow (BM) into the peripheral blood stream and the IV injection of an integrating, helper-dependent adenovirus (HD-Ad5/35++) vector system. These vectors target CD46, a receptor that is uniformly expressed on HSPCs. We demonstrated in human CD46 transgenic mice and immunodeficient mice with engrafted human CD34+ cells that HSPCs transduced in the periphery home back to the BM where they stably express the transgene. In hCD46 transgenic mice, we showed that our in vivo HSPC transduction approach allows for the stable transduction of primitive HSPCs. Twenty weeks after in vivo transduction, green fluorescent protein (GFP) marking in BM HSPCs (Lin-Sca1+Kit- cells) in most of the mice was in the range of 5% to 10%. The percentage of GFP-expressing primitive HSPCs capable of forming multilineage progenitor colonies (colony-forming units [CFUs]) increased from 4% of all CFUs at week 4 to 16% at week 12, indicating transduction and expansion of long-term surviving HSPCs. Our approach was well tolerated, did not result in significant transduction of nonhematopoietic tissues, and was not associated with genotoxicty. The ability to stably genetically modify HSPCs without the need of myeloablative conditioning is relevant for a broader clinical application of gene therapy.


Asunto(s)
Terapia Genética/métodos , Movilización de Célula Madre Hematopoyética/métodos , Proteína Cofactora de Membrana/biosíntesis , Transducción Genética/métodos , Adenoviridae , Animales , Vectores Genéticos/administración & dosificación , Células Madre Hematopoyéticas , Xenoinjertos , Humanos , Inyecciones Intravenosas , Ratones , Ratones Endogámicos C57BL
8.
Virus Genes ; 53(5): 684-691, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28593513

RESUMEN

Helper-dependent adenoviral vectors (HDAd) are deleted of all viral genes and they can efficiently transduce a wide variety of dividing and non-dividing cells to mediate high transgene expression levels. Unlike early generation adenoviral vectors, the absence of viral genes in HDAd results in long-term transgene expression without chronic toxicity and permits a large cloning capacity of 36 kb. Moreover, HDAd genomes exist extra-chromosomally thus minimizing the risks of germline transmission and insertional mutagenesis. For these reasons, HDAd offers tremendous potential for in vivo gene therapy. This chapter reviews preclinical studies using HDAd in large animal models to assess safety and efficacy in a wide variety of gene therapy applications.


Asunto(s)
Adenoviridae/genética , Genes Virales/genética , Vectores Genéticos/genética , Virus Helper/genética , Animales , Terapia Genética/métodos , Humanos , Modelos Animales , Transgenes/genética
9.
Mol Ther ; 21(4): 767-74, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23358188

RESUMEN

Helper-dependent adenoviral (HDAd) vectors can mediate long-term, high-level transgene expression from transduced hepatocytes with no chronic toxicity. However, a toxic acute response with potentially lethal consequences has hindered their clinical applications. Liver sinusoidal endothelial cells (LSECs) and Kupffer cells are major barriers to efficient hepatocyte transduction. Understanding the mechanisms of adenoviral vector uptake by non-parenchymal cells may allow the development of strategies aimed at overcoming these important barriers and to achieve preferential hepatocyte gene transfer with reduced toxicity. Scavenger receptors on Kupffer cells bind adenoviral particles and remove them from the circulation, thus preventing hepatocyte transduction. In the present study, we show that HDAd particles interact in vitro and in vivo with scavenger receptor-A (SR-A) and with scavenger receptor expressed on endothelial cells-I (SREC-I) and we exploited this knowledge to increase the efficiency of hepatocyte transduction by HDAd vectors in vivo through blocking of SR-A and SREC-I with specific fragments antigen-binding (Fabs).


Asunto(s)
Adenoviridae/genética , Receptor de Asialoglicoproteína/genética , Vectores Genéticos/genética , Receptores Inmunológicos/genética , Receptores Depuradores de Clase A/genética , Receptores Depuradores de Clase F/genética , Animales , Línea Celular , Técnica del Anticuerpo Fluorescente , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
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
11.
Hum Mol Genet ; 20(R1): R7-13, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21470977

RESUMEN

Helper-dependent adenoviral (HDAd) vectors devoid of all viral-coding sequences are promising non-integrating vectors for liver-directed gene therapy because they have a large cloning capacity, can efficiently transduce a wide variety of cell types from various species independent of the cell cycle and can result in long-term transgene expression without chronic toxicity. The main obstacle preventing clinical applications of HDAd for liver-directed gene therapy is the host innate inflammatory response against the vector capsid proteins that occurs shortly after intravascular vector administration resulting in acute toxicity, the severity of which is dependent on vector dose. Intense efforts have been focused on elucidating the factors involved in this acute response and various strategies have been investigated to improve the therapeutic index of HDAd vectors. These strategies have yielded encouraging results with the potential for clinical translation.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Virus Helper/genética , Hepatopatías/terapia , Hígado/citología , Animales , Vectores Genéticos/genética , Hepatocitos/metabolismo , Humanos , Inmunidad Innata , Hígado/metabolismo , Transducción Genética , Transgenes
12.
Toxicol Appl Pharmacol ; 268(3): 318-30, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23403069

RESUMEN

Adenoviral vectors (Ads) are promising gene delivery vehicles due to their high transduction efficiency; however, their clinical usefulness has been hampered by their immunogenicity and the presence of anti-Ad immunity in humans. We reported the efficacy of a gene therapy approach for glioma consisting of intratumoral injection of Ads encoding conditionally cytotoxic herpes simplex type 1 thymidine kinase (Ad-TK) and the immunostimulatory cytokine fms-like tyrosine kinase ligand 3 (Ad-Flt3L). Herein, we report the biodistribution, efficacy, and neurological and systemic effects of a bicistronic high-capacity Ad, i.e., HC-Ad-TK/TetOn-Flt3L. HC-Ads elicit sustained transgene expression, even in the presence of anti-Ad immunity, and can encode large therapeutic cassettes, including regulatory elements to enable turning gene expression "on" or "off" according to clinical need. The inclusion of two therapeutic transgenes within a single vector enables a reduction of the total vector load without adversely impacting efficacy. Because clinically the vectors will be delivered into the surgical cavity, normal regions of the brain parenchyma are likely to be transduced. Thus, we assessed any potential toxicities elicited by escalating doses of HC-Ad-TK/TetOn-Flt3L (1×10(8), 1×10(9), or 1×10(10) viral particles [vp]) delivered into the rat brain parenchyma. We assessed neuropathology, biodistribution, transgene expression, systemic toxicity, and behavioral impact at acute and chronic time points. The results indicate that doses up to 1×10(9) vp of HC-Ad-TK/TetOn-Flt3L can be safely delivered into the normal rat brain and underpin further developments for its implementation in a phase I clinical trial for glioma.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Ensayos Clínicos Fase I como Asunto/métodos , Citotoxinas/administración & dosificación , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Glioblastoma/tratamiento farmacológico , Inmunización/métodos , Adenoviridae/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Citotoxinas/efectos adversos , Citotoxinas/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Quimioterapia Combinada , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Vectores Genéticos/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Masculino , Ratas , Ratas Endogámicas Lew , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología , Resultado del Tratamiento
13.
Mol Ther ; 20(10): 1863-70, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22828499

RESUMEN

Hemophilia B is an excellent candidate for gene therapy because low levels of factor IX (FIX) (≥1%) result in clinically significant improvement of the bleeding diathesis. Helper-dependent adenoviral (HDAd) vectors can mediate long-term transgene expression without chronic toxicity. To determine the potential for HDAd-mediated liver-directed hemophilia B gene therapy, we administered an HDAd expressing hFIX into rhesus macaques through a novel and minimally invasive balloon occlusion catheter-based method that permits preferential, high-efficiency hepatocyte transduction with low, subtoxic vector doses. Animals given 1 × 10(12) and 1 × 10(11) virus particle (vp)/kg achieved therapeutic hFIX levels for the entire observation period (up to 1,029 days). At 3 × 10(10) and 1 × 10(10) vp/kg, only subtherapeutic hFIX levels were achieved which were not sustained long-term. Balloon occlusion administration of HDAd was well tolerated with negligible toxicity. Five of six animals developed inhibitors to hFIX. These results provide important information in assessing the clinical utility of HDAd for hemophilia B gene therapy.


Asunto(s)
Adenoviridae/genética , Cateterismo/métodos , Factor IX/genética , Vectores Genéticos , Hemofilia B/terapia , Macaca mulatta/genética , Adenoviridae/metabolismo , Animales , Modelos Animales de Enfermedad , Factor IX/metabolismo , Regulación de la Expresión Génica , Terapia Genética , Virus Helper/genética , Virus Helper/metabolismo , Hemofilia B/genética , Hígado/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción Genética/métodos , Transgenes/genética
14.
J Clin Invest ; 133(1)2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36301669

RESUMEN

Signaling circuits crucial to systemic physiology are widespread, yet uncovering their molecular underpinnings remains a barrier to understanding the etiology of many metabolic disorders. Here, we identified a copper-linked signaling circuit activated by disruption of mitochondrial function in the murine liver or heart that resulted in atrophy of the spleen and thymus and caused a peripheral white blood cell deficiency. We demonstrated that the leukopenia was caused by α-fetoprotein, which required copper and the cell surface receptor CCR5 to promote white blood cell death. We further showed that α-fetoprotein expression was upregulated in several cell types upon inhibition of oxidative phosphorylation. Collectively, our data argue that α-fetoprotein may be secreted by bioenergetically stressed tissue to suppress the immune system, an effect that may explain the recurrent or chronic infections that are observed in a subset of mitochondrial diseases or in other disorders with secondary mitochondrial dysfunction.


Asunto(s)
Cobre , Enfermedades Mitocondriales , Ratones , Animales , Cobre/metabolismo , alfa-Fetoproteínas/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Terapia de Inmunosupresión
15.
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
16.
Mol Ther ; 19(7): 1254-62, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21505422

RESUMEN

As much as 90% of an intravenously (i.v.) injected dose of adenovirus serotype 5 (Ad5) is absorbed and destroyed by liver Kupffer cells. Viruses that escape these cells can then transduce hepatocytes after binding factor X (FX). Given that interactions with FX and Kupffer cells are thought to occur on the Ad5 hexon protein, we replaced its exposed hypervariable regions (HVR) with those from Ad6. When tested in vivo in BALB/c mice and in hamsters, the Ad5/6 chimera mediated >10 times higher transduction in the liver. This effect was not due to changes in FX binding. Rather, Ad5/6 appeared to escape Kupffer cell uptake as evidenced by producing no Kupffer cell death in vivo, not requiring predosing in vivo, and being phagocytosed less efficiently by macrophages in vitro compared to Ad5. When tested as a helper-dependent adenovirus (Ad) vector, Ad5/6 mediated higher luciferase and factor IX transgene expression than either helper-dependent adenoviral 5 (HD-Ad5) or HD-Ad6 vectors. These data suggest that the Ad5/6 hexon-chimera evades Kupffer cells and may have utility for systemic and liver-directed therapies.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/genética , Macrófagos del Hígado/metabolismo , Transducción Genética/métodos , Alanina Transaminasa/sangre , Animales , Línea Celular , Cricetinae , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunohistoquímica , Interleucina-6/sangre , Macrófagos del Hígado/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa
17.
Mol Ther ; 19(1): 83-92, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20959811

RESUMEN

In vivo gene transfer with adenovirus vectors would significantly benefit from a tight control of the adenovirus-inherent liver tropism. For efficient hepatocyte transduction, adenovirus vectors need to evade from Kupffer cell scavenging while delivery to peripheral tissues or tumors could be improved if both scavenging by Kupffer cells and uptake by hepatocytes were blocked. Here, we provide evidence that a single point mutation in the hexon capsomere designed to enable defined chemical capsid modifications may permit both detargeting from and targeting to hepatocytes with evasion from Kupffer cell scavenging. Vector particles modified with small polyethylene glycol (PEG) moieties specifically on hexon exhibited decreased transduction of hepatocytes by shielding from blood coagulation factor binding. Vector particles modified with transferrin or, surprisingly, 5,000 Da PEG or dextran increased hepatocyte transduction up to 18-fold independent of the presence of Kupffer cells. We further show that our strategy can be used to target high-capacity adenovirus vectors to hepatocytes emphasizing the potential for therapeutic liver-directed gene transfer. Our approach may lead to a detailed understanding of the interactions between adenovirus vectors and Kupffer cells, one of the most important barriers for adenovirus-mediated gene delivery.


Asunto(s)
Adenoviridae/fisiología , Proteínas de la Cápside/genética , Técnicas de Transferencia de Gen , Hepatocitos/virología , Macrófagos del Hígado/virología , Hígado/virología , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Factores de Coagulación Sanguínea/metabolismo , Cápside/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Línea Celular Tumoral , Dextranos/metabolismo , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Células Hep G2 , Hepatocitos/metabolismo , Hepatocitos/fisiología , Humanos , Macrófagos del Hígado/metabolismo , Macrófagos del Hígado/fisiología , Hígado/metabolismo , Ratones , Ratones Endogámicos BALB C , Mutación Puntual , Polietilenglicoles/química , Transducción Genética/métodos , Transferrina/metabolismo , Tropismo/fisiología
18.
J Bronchology Interv Pulmonol ; 29(2): 125-130, 2022 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-34347696

RESUMEN

BACKGROUND: Coronavirus disease 2019 (COVID-19) can lead to hypoxemic respiratory failure resulting in prolonged mechanical ventilation. Typically, tracheostomy is considered in patients who remain ventilator dependent beyond 2 weeks. However, in the setting of this novel respiratory virus, the safety and benefits of tracheostomy are not well-defined. Our aim is to describe our experience with percutaneous tracheostomy in patients with COVID-19. MATERIALS AND METHODS: This is a single center retrospective descriptive study. We reviewed comorbidities and outcomes in patients with respiratory failure due to COVID-19 who underwent percutaneous tracheostomy at our institution from April 2020 to September 2020. In addition, we provide details of our attempt to minimize aerosolization by using a modified protocol with brief periods of planned apnea. RESULTS: A total of 24 patients underwent percutaneous tracheostomy during the study. The average body mass index was 33.0±10.0. At 30 days posttracheostomy 17 (71%) patients still had the tracheostomy tube and 14 (58%) remained ventilator dependent. There were 3 (13%) who died within 30 days. At the time of data analysis in November 2020, 9 (38%) patients had died and 7 (29%) had been decannulated. None of the providers who participated in the procedure experienced signs or symptoms of COVID-19 infection. CONCLUSION: Percutaneous tracheostomy in prolonged respiratory failure due to COVID-19 appears to be safe to perform at the bedside for both the patient and health care providers in the appropriate clinical context. Morbid obesity did not limit the ability to perform percutaneous tracheostomy in COVID-19 patients.


Asunto(s)
COVID-19 , Insuficiencia Respiratoria , COVID-19/complicaciones , Humanos , Insuficiencia Respiratoria/etiología , Insuficiencia Respiratoria/terapia , Estudios Retrospectivos , SARS-CoV-2 , Traqueostomía/efectos adversos , Traqueostomía/métodos
19.
Simul Healthc ; 2022 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-36378597

RESUMEN

INTRODUCTION: Performance assessment and feedback are critical factors in successful medical simulation-based training. The Dynamic Haptic Robotic Trainer (DHRT) allows residents to practice ultrasound-guided needle insertions during simulated central venous catheterization (CVC) procedures while providing detailed feedback and assessment. A study was performed to examine the effectiveness of the DHRT in training the important skills of needle tip tracking and aspiration and how these skills impact procedural complications in simulated CVC. METHODS: The DHRT data were collected for 163 residents at 2 hospitals for 6 simulated needle insertions. Users were given automated feedback on 5 performance metrics, which measure aspiration rate, arterial punctures, punctures through and through the vein, loss of access to the vein, and successful access to the vein. Aspiration rates and tip tracking rates were analyzed to determine their significance in preventing CVC complications and improving performance. RESULTS: Tip tracking rates higher than 40% were 2.3 times more likely to result in successful venous access than rates less than 10%. Similarly, aspiration rates higher than 80% were 2.6 times more likely to result in successful venous access than rates less than 10%. Proper tip tracking and aspiration both reduced mechanical complications. Resident performance improved for all metrics except tip tracking. CONCLUSIONS: Proper tip tracking and aspiration both reduced complications and increased the likelihood of success. However, the skill of tip tracking was not effectively learned through practice without feedback. Therefore, ultrasound-guided needle-based procedures, including CVC, can be improved by providing specific feedback to users on their ultrasound usage to track needle insertions.

20.
Am J Surg ; 224(3): 903-907, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-34930583

RESUMEN

BACKGROUND: This study compares surgical residents' knowledge acquisition of ultrasound-guided Internal Jugular Central Venous Catheterization (US-IJCVC) between in-person and online procedural training cohorts before receiving independent in-person Dynamic Haptic Robotic Simulation training. METHODS: Three surgical residency procedural training cohorts, two in-person (N = 26) and one online (N = 14), were compared based on their performance on a 24-item US-IJCVC evaluation checklist completed by an expert physician completed after training. Pre- and post-training US-IJCVC knowledge was also compared for the online cohort. RESULTS: No significant change in the pass rates on the US-IJCVC checklist was found between in-person and online cohorts (p = 0.208). There were differences in the Economy of Time and Motion between in-person and online cohorts (p < 0.005). The online cohort had significant increases in US-IJCVC knowledge pre-to post-training (p < 0.008). CONCLUSION: Online training with independent simulation practice was as effective as in-person training for US-IJCVC.


Asunto(s)
Cateterismo Venoso Central , Internado y Residencia , Entrenamiento Simulado , Competencia Clínica , Educación de Postgrado en Medicina , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA