Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Transfusion ; 60(5): 932-939, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32358832

RESUMEN

BACKGROUND: Hemoglobin-Based Oxygen Carriers (HBOCs) can act as an "oxygen bridge" in acute severe anemia when transfusion is indicated, but not possible. We present data on 10 Expanded Access (EA) patients treated with high cumulative doses of Hemopure (HBOC-201), to assess the ability of HBOC-201 to safely treat life threatening anemia in situations where high volumes of product were administered over an extended period of time. STUDY DESIGN AND METHODS: Inclusion in this study required that the patient receive at least 10 units of HBOC-201 between 2014 and 2017 under the FDA-sanctioned EA program. Depending on a patient's geographical location, treatment with HBOC-201 was obtained through either a single patient emergency Investigational New Drug (IND) application, or an intermediate size population IND. Of the 41 patients who were treated during this period, 10 patients received 10 or more units of the product. Data were obtained from medical records. RESULTS: Treatments with HBOC-201 started within 24 hours of signing consent and were administered at an average rate of 1.99 (SD 0.17) units per day over a mean of 8.2 days (SD 2.9), during which patients received on average 16.2 units (SD 5.7 units) of HBOC-201. The median pre-treatment nadir corpuscular hemoglobin (Hb) concentration was 3.3 (SD 0.9) g/dL and post-treatment Hemoglobin was 7.3 (SD 1.7) g/dL. Common side effects included methemoglobinemia, gastrointestinal symptoms, and hypertension. However, no product-related serious adverse events (SAEs) were noted. All patients survived. CONCLUSIONS: Administration of HBOC-201 over an extended period is a feasible and safe oxygen bridge for severely anemic patients who cannot be transfused with RBC.


Asunto(s)
Anemia/tratamiento farmacológico , Transfusión Sanguínea , Contraindicaciones , Hemoglobinas/administración & dosificación , Adulto , Anciano , Anemia/diagnóstico , Anemia/patología , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Hemoglobinas/efectos adversos , Humanos , Cuidados a Largo Plazo/métodos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Factores de Tiempo , Reacción a la Transfusión/prevención & control , Resultado del Tratamiento , Adulto Joven
2.
Mol Ther ; 17(12): 2115-20, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19809403

RESUMEN

Somatic cell gene transfer has permitted inducible gene expression in vivo through coinfection of multiple viruses. We hypothesized that the highly efficient plasmid-based piggyBac transposon system would enable long-term inducible gene expression in mice in vivo. We used a multiple-transposon delivery strategy to create a tetracycline-inducible expression system in vitro in human cells by delivering the two genes on separate transposons for inducible reporter gene expression along with a separate selectable transposon marker. Evaluation of stable cell lines revealed 100% of selected clones exhibited inducible expression via stable expression from three separate transposons simultaneously. We next tested and found that piggyBac-mediated gene transfer to liver or lung could achieve stable reporter gene expression in mice in vivo in either immunocompetent or immune deficient animals. A single injection of piggyBac transposons could achieve long-term inducible gene expression in the livers of mice in vivo, confirming our multiple-transposon strategy used in cultured cells. The plasmid-based piggyBac transposon system enables constitutive or inducible gene expression in vivo for potential therapeutic and biological applications without using viral vectors.


Asunto(s)
Elementos Transponibles de ADN/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Proteínas Fluorescentes Verdes/metabolismo , Transgenes/genética , Animales , Southern Blotting , Células Cultivadas , Femenino , Proteínas Fluorescentes Verdes/genética , Humanos , Riñón/citología , Riñón/metabolismo , Hígado/citología , Hígado/metabolismo , Hígado/virología , Pulmón/citología , Pulmón/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Integración Viral
3.
Mol Biol Cell ; 16(3): 1142-51, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15635102

RESUMEN

Peripheral myelin protein 22 (PMP22) is associated with a subset of hereditary peripheral neuropathies. Although predominantly recognized as a transmembrane constituent of peripheral nerve myelin, PMP22 is localized to epithelial and endothelial cell-cell junctions, where its function remains unknown. In this report, we investigated the role of PMP22 in epithelial biology. Expression of human PMP22 (hPMP22) slows cell growth and induces a flattened morphology in Madin-Darby canine kidney (MDCK) cells. The transepithelial electrical resistance (TER) and paracellular flux of MDCK monolayers are elevated by hPMP22 expression. After calcium switch, peptides corresponding to the second, but not the first, extracellular loop of PMP22 perturb the recovery of TER and paracellular flux. Finally, subsequent to wounding, epithelial monolayers expressing hPMP22 fail to migrate normally. These results indicate that PMP22 is capable of modulating several aspects of epithelial cell biology, including junctional permeability and wound closure.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/patología , Proteínas de la Mielina/metabolismo , Animales , Biotinilación , Western Blotting , Calcio/metabolismo , Comunicación Celular , Línea Celular , Movimiento Celular , Proliferación Celular , Perros , Impedancia Eléctrica , Epitelio/metabolismo , Epitelio/patología , Fase G1 , Crecimiento , Uniones Intercelulares/metabolismo , Microscopía Fluorescente , Vaina de Mielina/metabolismo , Péptidos/química , Permeabilidad , Fase de Descanso del Ciclo Celular , Fase S , Transgenes , Cicatrización de Heridas
4.
Cancer Res ; 78(2): 489-500, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29183891

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive disease lacking targeted therapy. In this study, we developed a CAR T cell-based immunotherapeutic strategy to target TEM8, a marker initially defined on endothelial cells in colon tumors that was discovered recently to be upregulated in TNBC. CAR T cells were developed that upon specific recognition of TEM8 secreted immunostimulatory cytokines and killed tumor endothelial cells as well as TEM8-positive TNBC cells. Notably, the TEM8 CAR T cells targeted breast cancer stem-like cells, offsetting the formation of mammospheres relative to nontransduced T cells. Adoptive transfer of TEM8 CAR T cells induced regression of established, localized patient-derived xenograft tumors, as well as lung metastatic TNBC cell line-derived xenograft tumors, by both killing TEM8+ TNBC tumor cells and targeting the tumor endothelium to block tumor neovascularization. Our findings offer a preclinical proof of concept for immunotherapeutic targeting of TEM8 as a strategy to treat TNBC.Significance: These findings offer a preclinical proof of concept for immunotherapeutic targeting of an endothelial antigen that is overexpressed in triple-negative breast cancer and the associated tumor vasculature. Cancer Res; 78(2); 489-500. ©2017 AACR.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Inmunoterapia , Neoplasias Pulmonares/terapia , Proteínas de Neoplasias/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Superficie Celular/metabolismo , Linfocitos T/trasplante , Neoplasias de la Mama Triple Negativas/terapia , Animales , Apoptosis , Biomarcadores de Tumor , Estudios de Casos y Controles , Proliferación Celular , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Ratones , Proteínas de Microfilamentos , Pronóstico , Tasa de Supervivencia , Linfocitos T/inmunología , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
FASEB J ; 20(14): 2594-6, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17065228

RESUMEN

Pulmonary hypertension (PH) is a life-threatening disorder with high mortality rates and limited treatment options. Gene therapy is an alternative treatment strategy, yet viral vectors have inherent disadvantages including immune activation. The Sleeping Beauty (SB) transposon is a nonviral method of gene delivery that overcomes some of these drawbacks. A SB-based transposon harboring a constitutively active endothelial nitric oxide synthase (eNOS) gene was administered to Sprague-Dawley rats via tail vein injection using the carrier polyethylenimine. Two days after transposon delivery, monocrotaline (MCT) was administered to induce PH. Hemodynamic, histological, and molecular measurements were performed four weeks later. Animals coinjected with transposase showed a significant reduction in pulmonary arterial pressure (PABP, 31.67+/-6.03 mmHg, P<0.01), an attenuation of right ventricle (RV) to whole heart (WH) wt ratios (0.227+/-0.0252, P<0.05) and a decrease in the pulmonary vessel wall thickness index (36.87%, P<0.001), compared with those animals receiving the eNOS transposon and a nonfunctional transposase (PABP 44.33+/-4.04 mmHg; RV/WH ratio 0.280+/-0.01; wall thickness index 62.14%) or control animals receiving MCT injection alone (PABP 49.67+/-3.22 mmHg; RV/WH ratio 0.290+/-0.0265; wall thickness index 71.99%). The physiological improvements correlated with therapeutic gene expression, suggesting that transposon-based genetic approaches have utility in the treatment of PH.


Asunto(s)
Elementos Transponibles de ADN , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/terapia , Monocrotalina/toxicidad , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Regulación Enzimológica de la Expresión Génica , Masculino , Óxido Nítrico Sintasa de Tipo III/genética , Ratas , Ratas Sprague-Dawley
6.
FASEB J ; 20(13): 2384-6, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17015408

RESUMEN

Sleeping Beauty (SB) transposon is a natural nonviral gene transfer system that can mediate long-term transgene expression. Its potential utility in treating organ transplantation-associated long-term complications has not yet been explored. In the present study we generated an improved SB transposon encoding the human gene indoleamine-2,3-dioxygenase (hIDO), an enzyme that possesses both T cell-suppressive and antioxidant properties and selectively delivered the SB transposon in combination with a hyperactive transposase plasmid to donor lung using the cationic polymer polyethylenimine (PEI) as transfection reagent. This nonviral gene therapeutic approach led to persistent and uniform transgene expression in the rat lung tissue without noticeable toxicity and inflammation. Importantly, IDO activity produced by hIDO transgene showed a remarkable therapeutic response, as evident by near normal pulmonary function (peak airway pressure and oxygenation), histological appearance, and reduced collagen content in lung allografts. In addition, we established a hIDO-overexpressing type II cell line using the SB-based gene transfer system and found that hIDO-overexpressing lung cells effectively inhibited transforming growth factor-beta-stimulated fibroblast proliferation in vitro. In summary, the SB-based gene therapy with hIDO represents a new strategy for treating lung transplantation-associated chronic complications, e.g., obliterative bronchiolitis.


Asunto(s)
Fibrosis/prevención & control , Terapia Genética/métodos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/uso terapéutico , Neoplasias Pulmonares/patología , Transposasas/genética , Animales , Animales Modificados Genéticamente , Elementos Transponibles de ADN , Humanos , Ratas , Trasplante Homólogo/patología
7.
BioDrugs ; 20(4): 219-29, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16831021

RESUMEN

Safe and effective delivery of genetic material to mammalian tissues would significantly expand the therapeutic possibilities for a large number of medical conditions. Unfortunately, the promise of gene therapy has been hampered by technical challenges, the induction of immune responses, and inadequate expression over time. Despite these setbacks, progress continues to be made and the anticipated benefits may come to fruition for certain disorders. In terms of delivery, nonviral vector systems are particularly attractive as they are simple to produce, can be stored for long periods of time, and induce no specific immune responses. A significant drawback to nonviral systems has been the lack of persistent expression, as plasmids are lost or degraded when delivered to living tissues. The recent application of integrating transposons to nonviral gene delivery has significantly helped to overcome this obstacle, because it allows for genomic integration and long-term expression. Recent advances in transposon-based vector systems hold promise as new technologies that may unlock the potential of gene therapy; however, technical and safety issues still need refinement.


Asunto(s)
Elementos Transponibles de ADN/genética , Terapia Genética/métodos , Transposasas/uso terapéutico , Animales , Marcación de Gen/métodos , Técnicas de Transferencia de Gen , Humanos , Modelos Biológicos , Riesgo
8.
Biochim Biophys Acta ; 1570(1): 53-62, 2002 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11960689

RESUMEN

We have previously shown that glucose deprivation activates glucose transport in a time- and protein synthesis-dependent fashion in 3T3-L1 adipocytes, a mouse cell line. Coincident with this is loss of glycogen. Because glycogen phosphorylase (GP) is responsible for glycogen degradation, we have examined its regulation to determine the relationship between transport activation and glycogen turnover. We first cloned the adipose GP cDNA and found sequence similarity to rat and human liver GP. Because the mouse liver GP cDNA sequence was unavailable, we cloned this cDNA as well and showed 100% identity between mouse adipose and liver sequences. A 3.1 kb transcript was readily observed in total RNA isolated from mouse liver or adipose by Northern blot analysis but, surprisingly, not in either total or poly(A) selected RNA from 3T3-L1 adipocytes. To evaluate regulation in 3T3-L1 adipocytes, we amplified GP mRNA from total RNA using multiplex, semi-quantitative PCR but found that expression did not change in response to deprivation. GP protein levels did not change either. However, endogenous GP activity from glucose-deprived cells was significantly elevated relative to controls, due to an increase in the phosphorylated form of GP (GPa). Finally, we overexpressed GP to determine its direct influence on the glucose transport system. These results were negative, which suggests that the nutrient control of glucose transport and GP occurs independently despite kinetic similarities in transport activation and glycogen turnover.


Asunto(s)
Adipocitos/metabolismo , Glucosa/deficiencia , Glucógeno Fosforilasa/metabolismo , Células 3T3 , Adipocitos/enzimología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Transporte Biológico , Clonación Molecular , ADN Complementario/biosíntesis , ADN Complementario/química , Activación Enzimática , Regulación de la Expresión Génica , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1 , Glucógeno Fosforilasa/biosíntesis , Glucógeno Fosforilasa/genética , Isoenzimas/biosíntesis , Hígado/enzimología , Ratones , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
9.
Anticancer Res ; 31(10): 3417-22, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21965755

RESUMEN

BACKGROUND/AIM: Tumor endothelial marker 8 (TEM8) is a tumor endothelial-associated antigen that is having an increasingly recognized role in tumor biology. The expression of TEM8 in triple-negative breast cancer (TNBC) has not yet been characterized. MATERIALS AND METHODS: We hypothesize that TEM8 is overexpressed in TNBC and in metastatic TNBC in lymph nodes (LN) compared to normal breast tissue and normal lymphatic tissue, respectively. We studied expression of TEM8 in cases of primary (n=17) and metastatic (n=2) TNBC using immunohistochemical analyses. RESULTS: All cases demonstrated increased expression of TEM8 in tumor tissue compared to non-cancerous breast tissue. TEM8 was expressed at a higher level in the stroma adjacent to the TNBC in all cases, with focal immunoreactive areas within the tumor. TEM8 was not expressed in normal lymphoid tissue, but showed expression at sites of LN metastases. CONCLUSION: TEM8 would appear to represent a new biologic target for designing novel diagnostic or therapeutic approaches for TNBC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superficie Celular/metabolismo , Anticuerpos Antineoplásicos/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Femenino , Humanos , Inmunohistoquímica , Ganglios Linfáticos/patología , Proteínas de Microfilamentos , Proteínas de Neoplasias/inmunología , Receptores de Superficie Celular/inmunología
10.
Cold Spring Harb Protoc ; 2010(11): pdb.prot5521, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21041394

RESUMEN

RNA interference (RNAi) is an evolutionarily conserved process that silences gene expression through double-stranded RNA species in a sequence-specific manner. Small interfering RNAs (siRNAs) can promote sequence-specific degradation and/or translational repression of target RNA by activation of the RNA-induced silencing complex (RISC). Traditionally, silencing in mammalian cells had been achieved by transfection of synthetically derived siRNA duplexes, resulting in transient gene suppression of the target sequence. As the technology was advanced, inhibitory short-hairpin-shaped RNAs (shRNAs) could be produced by transcription from RNA polymerase-III (pol-III)-driven promoters, such as H1, U6, or cytomegalovirus (CMV)-enhanced pol III promoters. Following transcription, the shRNAs are processed by the enzyme Dicer into active siRNA. This approach allows for the continuous production of siRNA within cells using a DNA template and offers increased options for delivery of the pol-III-driven transcriptional units. A number of different viral vectors, as well as plasmid DNAs, have been utilized to deliver shRNA to mammalian cells. Here, the Tc1/mariner DNA transposon Sleeping Beauty (SB) is used as a tool to deliver shRNA-encoding transcriptional units. The SB transposon system uses a "cut-and-paste" mechanism to insert the transposon into random TA dinucleotides within the target genome. The shRNAs are then processed and used for gene knockdown.


Asunto(s)
Elementos Transponibles de ADN , Células Eucariotas , Técnicas de Transferencia de Gen , Biología Molecular/métodos , ARN Interferente Pequeño/genética , Animales , Humanos , Mamíferos
11.
Cancer Res ; 69(12): 5126-32, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19528090

RESUMEN

Tumor endothelial marker 8 (TEM8) is a recently described protein that is preferentially expressed within tumor endothelium. We have developed a fusion protein that targets TEM8 and disrupts tumor vasculature by promoting localized thrombosis. Fusion protein specificity and function were evaluated using Western blot analysis, ELISA, and enzymatic assays. A xenograft model of colorectal carcinoma was used to test the efficacy of targeted and control fusion proteins. Mice treated with the gene encoding anti-TEM8/truncated tissue factor exhibited a 53% reduction in tumor volume when compared with the untreated animals (P < 0.0001; n = 10) and achieved a 49% increase in tumor growth delay by Kaplan-Meier analysis (P = 0.0367; n = 6). Immunohistochemistry confirmed tumor endothelial expression of TEM8, fusion protein homing to tumor vasculature, decrease in vessel density, and localized areas of thrombosis. These data support the hypothesis that targeting TEM8 can be an effective approach to influence tumor development by disrupting tumor vasculature.


Asunto(s)
Neoplasias Colorrectales/irrigación sanguínea , Receptores de Péptidos/metabolismo , Animales , Secuencia de Bases , Biomarcadores de Tumor , Western Blotting , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Microfilamentos , Receptores de Superficie Celular , Proteínas Recombinantes de Fusión/farmacología
12.
Microb Biotechnol ; 1(1): 62-7, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21261822

RESUMEN

The prokaryotic CpG-specific DNA methylase from Spiroplasma, SssI methylase, has been extensively used to methylate plasmid DNA in vitro to investigate the effects of methylation in vertebrate systems. Currently available methods to produce CpG-methylated plasmid DNA have certain limitations and cannot generate large quantities of methylated DNA without cost or problems of purity. Here we describe an approach in which the SssI methylase gene has been introduced into the Escherichia coli bacterial genome under the control of an inducible promoter. Plasmid DNA propagated in this bacterium under conditions which induce the methylase gene result in significant (>90%) CpG methylation. Methylated DNA produced by this approach behaves similarly to methylated DNA produced in vitro using the purified methylase. The approach is scalable allowing for the production of milligram quantities of methylated plasmid DNA.


Asunto(s)
Proteínas Bacterianas/metabolismo , Metilación de ADN , ADN Bacteriano/metabolismo , ADN-Citosina Metilasas/metabolismo , Escherichia coli/metabolismo , Ingeniería Genética/métodos , Plásmidos/metabolismo , Spiroplasma/enzimología , Proteínas Bacterianas/genética , ADN Bacteriano/genética , ADN-Citosina Metilasas/genética , Escherichia coli/genética , Expresión Génica , Plásmidos/genética , Spiroplasma/genética
13.
Mol Ther ; 13(5): 1006-15, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16464640

RESUMEN

Hemophilia A, deficiency of coagulation factor VIII (FVIII), is an attractive candidate for gene therapy as expression of modest amounts of FVIII can provide therapeutic benefit. Most gene transfer approaches for hemophilia have focused on the liver, as this is the major source of endogenous FVIII; however, increasing evidence suggests that endothelial cells are capable of synthesis and release of FVIII. Here the Sleeping Beauty (SB) transposon is employed to target long-term expression of the human B-domain-depleted FVIII gene (approved gene symbol F8) within lung endothelia of hemophilic mice. As the formation of inhibitory antibodies to FVIII has been a significant impediment toward achieving therapeutic plasma levels after gene or protein therapy, we chose to perform gene transfer in neonatal mice, which are more likely to be immune tolerant. Using this approach, low therapeutic levels of FVIII ( approximately 10%), as well as phenotypic correction of the bleeding disorder, were achieved in all animals that received the FVIII transposon and functional transposase throughout the duration of the study (24 weeks). Rechallenge of these animals with additional gene transfer did not result in significant increases in FVIII levels, due mainly to increases in inhibitory antibodies. These studies demonstrate the feasibility of using endothelial-targeted SB transposons for the treatment of hemophilia A.


Asunto(s)
Factor VIII/genética , Terapia Genética/métodos , Hemofilia A/genética , Hemofilia A/terapia , Transposasas/genética , Animales , Animales Recién Nacidos , Coagulación Sanguínea , Línea Celular , Endotelio Vascular/citología , Factor VIII/metabolismo , Estudios de Factibilidad , Expresión Génica , Técnicas de Transferencia de Gen , Hemofilia A/inmunología , Humanos , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo
14.
Am J Respir Crit Care Med ; 173(5): 566-72, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16293800

RESUMEN

RATIONALE: Lung allografts are prone to reperfusion injury and acute rejection, which, in addition to infiltrating lymphocytes, are accompanied by neutrophil infiltration and neutrophil-associated oxidative stress. Indoleamine 2,3-dioxygenase (IDO) is a unique cytosolic enzyme that possesses T-cell-suppressive and antioxidant properties. OBJECTIVES: The purpose of this study was to determine if genetic up-regulation of IDO could ameliorate acute lung allograft injury. METHODS: Lung orthotopic transplants were performed using Lewis donors and Sprague-Dawley rat recipients (allografts) or the same strain (isografts). Plasmid-encoding human IDO was delivered to donor lungs in vivo using a nonviral gene-transfer vector, polyethylenimine. Transplanted lungs were evaluated at 6 d post-transplantation based on pulmonary function, histology, inflammatory responses, and their associated oxidative stress. Basic biology of the IDO-overexpressing lung cells was evaluated in vitro in response to external oxidant. MEASUREMENTS AND MAIN RESULTS: This gene delivery method led to uniform transgene expression in lung tissue distributed in airway, alveolar epithelial, and endothelial cells. IDO overexpression in lung allografts resulted in a significant protective effect with improvement in functional properties (peak airway pressure and oxygenation) and histologic appearance. Although IDO was able to block local T-cell responses, it failed to abrogate neutrophilic infiltration and the inflammation-associated oxidative stress. IDO-enhanced lung cells were resistance to oxidant-induced necrosis and apoptosis by limiting intracellular reactive oxygen species formation. CONCLUSIONS: These results demonstrate that IDO prevents acute lung allograft injury through augmenting the local antioxidant defense system and inhibiting alloreactive T-cell responses.


Asunto(s)
Terapia Genética , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Trasplante de Pulmón/efectos adversos , Complicaciones Posoperatorias/prevención & control , Enfermedad Aguda , Animales , Masculino , Complicaciones Posoperatorias/inmunología , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley
15.
J Neurochem ; 92(6): 1531-41, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15748170

RESUMEN

Accumulation of misfolded proteins and alterations in the ubiquitin-proteasome pathway are associated with various neurodegenerative conditions of the CNS and PNS. Aggregates containing ubiquitin and peripheral myelin protein 22 (PMP22) have been observed in the Trembler J mouse model of Charcot-Marie-Tooth disease type 1A demyelinating neuropathy. In these nerves, the turnover rate of the newly synthesized PMP22 is reduced, suggesting proteasome impairment. Here we show evidence of proteasome impairment in Trembler J neuropathy samples compared with wild-type, as measured by reduced degradation of substrate reporters. Proteasome impairment correlates with increased levels of polyubiquitinated proteins, including PMP22, and the recruitment of E1, 20S and 11S to aggresomes formed either spontaneously due to the Trembler J mutation or upon proteasome inhibition. Furthermore, myelin basic protein, an endogenous Schwann cell proteasome substrate, associates with PMP22 aggregates in affected nerves. Together, our data show that in neuropathy nerves, reduced proteasome activity is coupled with the accumulation of ubiquitinated substrates, and the recruitment of proteasomal pathway constituents to aggregates. These results provide novel insights into the mechanism by which altered degradation of Schwann cell proteins may contribute to the pathogenesis of certain PMP22 neuropathies.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/enzimología , Proteínas de la Mielina/metabolismo , Nervios Periféricos/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Animales , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/fisiopatología , Modelos Animales de Enfermedad , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/metabolismo , Ratones , Ratones Mutantes Neurológicos , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Fibras Nerviosas Mielínicas/metabolismo , Fibras Nerviosas Mielínicas/patología , Nervios Periféricos/patología , Nervios Periféricos/fisiopatología , Complejo de la Endopetidasa Proteasomal/genética , Ratas , Células de Schwann/metabolismo
16.
Mol Ther ; 12(6): 1148-56, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16150650

RESUMEN

Transposable elements have enormous potential to overcome one of the major hurdles in nonviral gene delivery, namely the lack of long-term gene expression. The Sleeping Beauty (SB) transposon is a promising vector system for nonviral gene therapy as it has the highest transposition activity of all known DNA transposons within mammalian cells. In an effort to generate a more efficient delivery vehicle, we conducted a systematic evaluation of several novel and previously identified SB transposase mutants. The results indicate that certain combinations of mutants do not enhance transposition, whereas others give a synergistic response. The most active mutant, designated HSB17, shows nearly 17-fold higher transposition activity compared to the original transposase SB10 when tested within the same expression cassette. In addition, synergistic activity is observed when this hyperactive mutant is combined with an improved transposon. Animal studies utilizing the hyperactive transposase show enhanced long-term reporter gene expression. These modifications further expand the utility of this transposon-based gene transfer system.


Asunto(s)
Elementos Transponibles de ADN/genética , Terapia Genética/métodos , Vectores Genéticos , Transposasas/genética , Secuencia de Aminoácidos , Animales , Western Blotting , Codón , Técnicas de Transferencia de Gen , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Modelos Genéticos , Datos de Secuencia Molecular , Mutación , Filogenia , Plásmidos/metabolismo , Homología de Secuencia de Aminoácido , Transfección
17.
Genomics ; 85(6): 762-73, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15885502

RESUMEN

A cDNA fragment originally identified in U-937 cells as a vitamin D(3)-regulated gene is here designated the glutamate-rich WD-repeat (GRWD1) gene. WD-repeat proteins are a class of functionally divergent molecules that cooperate with other proteins to regulate cellular processes. GRWD1 encodes a 446-amino-acid protein containing a glutamate-rich region followed by four WD repeats. The yeast homologue of GRWD1, Rrb1, has been shown to be an essential protein involved in ribosome biogenesis. Northern analysis of GRWD1 message levels in the myeloid cell line HL-60 undergoing differentiation induced by vitamin D(3) or retinoic acid demonstrate downregulation coincident with slowing of cellular proliferation. A siRNA designed to downregulate GRWD1 similarly results in a decrease in cellular proliferation within 293 cells. Metabolic labeling of cells expressing the siRNA to GRWD1 shows a decrease in global protein synthesis. Finally, nuclear fractionation studies show cosedimentation of GRWD1 with preribosomal complexes, as well as the WD-repeat-containing protein Bop1, which has previously been implicated in ribosome biogenesis. These studies suggest that within mammalian cells GRWD1 plays a role in ribosome biogenesis and during myeloid differentiation its levels are regulated.


Asunto(s)
Proteínas Portadoras/metabolismo , Diferenciación Celular/efectos de los fármacos , Colecalciferol/farmacología , Ribosomas/metabolismo , Secuencia de Bases , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Células HL-60 , Humanos , Datos de Secuencia Molecular , Células U937
18.
Biochem Biophys Res Commun ; 316(3): 643-50, 2004 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-15033448

RESUMEN

RNA interference is rapidly becoming a powerful tool for genetic analyses in mammalian systems. A potential drawback to transient small inhibitory RNA silencing is the short duration of downregulation it confers, usually only 24-72h. Viral-based vector systems for the long-term delivery of RNA hairpins have been developed, yet they require expertise in viral production and transduction. Here we describe a simple plasmid-based system for the generation of long-term gene knockdown utilizing RNA interference combined with the gene delivery capabilities of the mammalian Tc1-like transposon Sleeping Beauty. Designated Maleficent, this system is shown to downregulate exogenous expression of GFP in a constitutively positive cell line. In addition, targeting of the endogenously expressed lamin A gene results in long-term silencing with significant reduction in protein levels (> 95%). Maleficent therefore provides a relatively easy, efficient, and stable means of delivering RNAi hairpins to generate long-term gene-specific knockdown cell lines.


Asunto(s)
Técnicas Genéticas , Interferencia de ARN , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Western Blotting , Línea Celular , Separación Celular , Pollos , Elementos Transponibles de ADN , Regulación hacia Abajo , Citometría de Flujo , Silenciador del Gen , Vectores Genéticos , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Lamina Tipo A/biosíntesis , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Datos de Secuencia Molecular , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo , Factores de Tiempo , Transfección
19.
Am J Physiol Endocrinol Metab ; 286(4): E568-76, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14665446

RESUMEN

Glucose deprivation dramatically increases glucose transport activity in 3T3-L1 adipocytes without changing the concentration of GLUT1 in the plasma membrane (PM). Recent data suggest that subcompartments within the PM, specifically lipid rafts, may sequester selected proteins and alter their activity. To evaluate this possibility, we examined the distribution of GLUT1 in Triton X-100-soluble and -insoluble fractions. Our data show that 77% of the GLUT1 pool in PMs isolated from control 3T3-L1 adipocytes was extracted by 0.2% Triton X-100. After glucose deprivation for 12 h, only 56% of GLUT1 was extracted by detergent. In contrast, there was a twofold increase in the GLUT1 content of the detergent-resistant fraction. To evaluate whether GLUT1 interacts with a specific protein within lipid rafts, we focused on stomatin, recently shown to interact with and inhibit GLUT1 activity. Stomatin is distributed about equally between the PM and the biosynthetic compartments, and its expression is not affected by glucose deprivation. Nearly 90% of the PM pool of stomatin is in detergent-resistant lipid rafts. In normal 3T3-L1 adipocytes, we were unable to demonstrate an interaction between GLUT1 and stomatin in coimmunoprecipitation experiments. However, in stomatin-overexpressing cells, there was clear coprecipitation of stomatin with GLUT1 antibodies. Glucose deprivation increased this interaction threefold, which may reflect the increase of GLUT1 in lipid rafts. Despite this, there was little change in transport activity in glucose-deprived, stomatin-overexpressing cells vs. that in control cells. Thus GLUT1 interacts with stomatin in lipid rafts, but this interaction per se does not alter transport activity. Rather, stomatin may serve as an anchor for GLUT1 in lipid rafts, the environment of which favors activation.


Asunto(s)
Adipocitos/metabolismo , Glucosa/fisiología , Microdominios de Membrana/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Células 3T3 , Animales , Proteínas Sanguíneas/antagonistas & inhibidores , Proteínas Sanguíneas/inmunología , Northern Blotting , Western Blotting , Línea Celular , Transportador de Glucosa de Tipo 1 , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Ratones , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo
20.
Mol Ther ; 10(1): 97-105, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15233946

RESUMEN

Endothelial cells have complex roles in the pathophysiology of vascular and heart disease and are increasingly being recognized as targets for gene therapy. The intravenous administration of plasmid DNA complexed to lipid tends to target transfection of endothelial cells within the lung; however, expression from the transgene remains transient. Here we utilize the integrating capability of the Sleeping Beauty (SB) transposon for durable gene transfer within lung endothelia. To restrict expression of the transgene, an endothelial cell-specific promoter, endothelin-1, was placed within the transposon. Further refinements to the transposon increased in vitro transposition efficiency by 3.6-fold. Utilizing this optimized transposon we evaluated the expression of two reporter molecules, secreted alkaline phosphatase (SEAP) and intracellular GFP, following administration of DNA-polyethylenimine complexes to mice. Long-term expression (>2 months) of SEAP occurred only with cotransfection of adequate amounts of transposase. Localization studies using the GFP reporter, at 3 days and 6 weeks postinjection, demonstrated that the majority of transgene-expressing cells were of endothelial origin, while the second most abundant cell type was type II pneumocyte. These results suggest that the SB transposon can be adapted to target particular cell types, in this case, endothelial cells. Such an approach may be useful for gene therapy paradigms involving the long-term modulation of vascular and endothelial cell biology.


Asunto(s)
Elementos Transponibles de ADN/genética , Endotelina-1/genética , Terapia Genética/métodos , Pulmón/metabolismo , Fosfatasa Alcalina/análisis , Fosfatasa Alcalina/genética , Animales , Línea Celular , Células Endoteliales/metabolismo , Genes Reporteros/genética , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Humanos , Pulmón/química , Pulmón/citología , Ratones , Ratones Endogámicos , Plásmidos/administración & dosificación , Plásmidos/química , Plásmidos/genética , Polietileneimina/química , Regiones Promotoras Genéticas/genética , Transposasas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA