Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Clin Exp Immunol ; 173(2): 230-41, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23607394

RESUMEN

Replication-deficient adenoviral (Ad) vectors of non-human serotypes can serve as Ad vaccine platforms to circumvent pre-existing anti-human Ad immunity. We found previously that, in addition to that feature, a non-human primate-based AdC7 vector expressing outer membrane protein F of P. aeruginosa (AdC7OprF) was more potent in inducing lung mucosal and protective immunity compared to a human Ad5-based vector. In this study we analysed if genetic modification of the AdC7 fibre to display an integrin-binding arginine-glycine-aspartic acid (RGD) sequence can further enhance lung mucosal immunogenicity of AdC7OprF. Intratracheal immunization of mice with either AdC7OprF.RGD or AdC7OprF induced robust serum levels of anti-OprF immunoglobulin (Ig)G up to 12 weeks that were higher compared to immunization with the human vectors Ad5OprF or Ad5OprF.RGD. OprF-specific cellular responses in lung T cells isolated from mice immunized with AdC7OprF.RGD and AdC7OprF were similar for T helper type 1 (Th1) [interferon (IFN)-γ in CD8(+) and interleukin (IL)-12 in CD4(+)], Th2 (IL-4, IL-5 and IL-13 in CD4(+)) and Th17 (IL-17 in CD4(+)). Interestingly, AdC7OprF.RGD induced more robust protective immunity against pulmonary infection with P. aeruginosa compared to AdC7OprF or the control Ad5 vectors. The enhanced protective immunity induced by AdC7OprF.RGD was maintained in the absence of alveolar macrophages (AM) or CD1d natural killer T cells. Together, the data suggest that addition of RGD to the fibre of an AdC7-based vaccine is useful to enhance its mucosal protective immunogenicity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Pulmón/inmunología , Oligopéptidos , Porinas/metabolismo , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Células TH1/inmunología , Adenoviridae/genética , Animales , Células Cultivadas , Citocinas/inmunología , Femenino , Vectores Genéticos/genética , Humanos , Inmunidad Mucosa , Inmunoglobulina G/sangre , Ratones , Ratones Endogámicos C57BL , Oligopéptidos/genética , Porinas/genética , Primates , Infecciones por Pseudomonas/prevención & control , Vacunas contra la Infección por Pseudomonas , Pseudomonas aeruginosa/genética
2.
Eur Respir J ; 38(1): 15-28, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21349909

RESUMEN

The glutathione S-transferase (GST) enzymes catalyse the conjugation of xenobiotics to glutathione. Based on reports that inherited copy number variations (CNVs) modulate some GST gene expression levels, and that the small airway epithelium (SAE) and alveolar macrophages (AMs) are involved early in the pathogenesis of smoking-induced lung disease, we asked: do germline CNVs modulate GST expression levels in SAE and AMs? Microarrays were used to survey GST gene expression and determine CNVs genotypes in SAE and AMs obtained by bronchoscopy from current smokers and nonsmokers. 26% of subjects were null for both GSTM1 alleles, with reduced GSTM1 mRNA levels seen in both SAE and AMs. 30% of subjects had homozygous deletions of GSTT1, with reduced mRNA levels in both tissues. Interestingly, GSTT2B exhibited homozygous deletion in the blood of 27% of subjects and was not expressed in SAE in the remainder of subjects, but was expressed in AMs of heterozygotes and wild-type subjects, proportionate to genotype. These data show a germline CNV-mediated linear relationship of genotype with expression level, suggesting minimal compensation of gene expression levels in heterozygotes, consistent with GST polymorphisms playing a role in the risk of smoking-associated, xenobiotic-induced lung disease.


Asunto(s)
Dosificación de Gen , Regulación de la Expresión Génica , Glutatión Transferasa/genética , Pulmón/metabolismo , Adulto , Alelos , Femenino , Perfilación de la Expresión Génica , Genotipo , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo Genético
3.
Nat Med ; 7(11): 1194-201, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11689883

RESUMEN

The role of bone marrow (BM)-derived precursor cells in tumor angiogenesis is not known. We demonstrate here that tumor angiogenesis is associated with recruitment of hematopoietic and circulating endothelial precursor cells (CEPs). We used the angiogenic defective, tumor resistant Id-mutant mice to show that transplantation of wild-type BM or vascular endothelial growth factor (VEGF)-mobilized stem cells restore tumor angiogenesis and growth. We detected donor-derived CEPs throughout the neovessels of tumors and Matrigel-plugs in an Id1+/-Id3-/- host, which were associated with VEGF-receptor-1-positive (VEGFR1+) myeloid cells. The angiogenic defect in Id-mutant mice was due to impaired VEGF-driven mobilization of VEGFR2+ CEPs and impaired proliferation and incorporation of VEGFR1+ cells. Although targeting of either VEGFR1 or VEGFR2 alone partially blocks the growth of tumors, inhibition of both VEGFR1 and VEGFR2 was necessary to completely ablate tumor growth. These data demonstrate that recruitment of VEGF-responsive BM-derived precursors is necessary and sufficient for tumor angiogenesis and suggest new clinical strategies to block tumor growth.


Asunto(s)
Células Madre Hematopoyéticas/patología , Proteínas de Neoplasias , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/patología , Neovascularización Patológica , Proteínas Represoras , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Endotelio Vascular/patología , Trasplante de Células Madre Hematopoyéticas , Proteína 1 Inhibidora de la Diferenciación , Proteínas Inhibidoras de la Diferenciación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Mutación , Neovascularización Patológica/genética , Pruebas de Neutralización , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Factores de Crecimiento/fisiología , Receptores de Factores de Crecimiento Endotelial Vascular , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular
4.
J Exp Med ; 193(9): 1005-14, 2001 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-11342585

RESUMEN

Tyrosine kinase receptors for angiogenic factors vascular endothelial growth factor (VEGF) and angiopoietin-1 (Ang-1) are expressed not only by endothelial cells but also by subsets of hematopoietic stem cells (HSCs). To further define their role in the regulation of postnatal hematopoiesis and vasculogenesis, VEGF and Ang-1 plasma levels were elevated by injecting recombinant protein or adenoviral vectors expressing soluble VEGF(165), matrix-bound VEGF(189), or Ang-1 into mice. VEGF(165), but not VEGF(189), induced a rapid mobilization of HSCs and VEGF receptor (VEGFR)2(+) circulating endothelial precursor cells (CEPs). In contrast, Ang-1 induced delayed mobilization of CEPs and HSCs. Combined sustained elevation of Ang-1 and VEGF(165) was associated with an induction of hematopoiesis and increased marrow cellularity followed by proliferation of capillaries and expansion of sinusoidal space. Concomitant to this vascular remodeling, there was a transient depletion of hematopoietic activity in the marrow, which was compensated by an increase in mobilization and recruitment of HSCs and CEPs to the spleen resulting in splenomegaly. Neutralizing monoclonal antibody to VEGFR2 completely inhibited VEGF(165), but not Ang-1-induced mobilization and splenomegaly. These data suggest that temporal and regional activation of VEGF/VEGFR2 and Ang-1/Tie-2 signaling pathways are critical for mobilization and recruitment of HSCs and CEPs and may play a role in the physiology of postnatal angiogenesis and hematopoiesis.


Asunto(s)
Factores de Crecimiento Endotelial/fisiología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Linfocinas/fisiología , Glicoproteínas de Membrana/fisiología , Proteínas Proto-Oncogénicas , Transducción de Señal/fisiología , Adenoviridae , Angiopoyetina 1 , Animales , Células de la Médula Ósea , Factores de Crecimiento Endotelial/administración & dosificación , Factores de Crecimiento Endotelial/sangre , Factores de Crecimiento Endotelial/metabolismo , Femenino , Vectores Genéticos , Movilización de Célula Madre Hematopoyética , Leucocitos/fisiología , Linfocinas/administración & dosificación , Linfocinas/sangre , Linfocinas/metabolismo , Masculino , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/sangre , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular , Bazo/citología , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
5.
J Clin Invest ; 101(8): 1789-96, 1998 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-9541511

RESUMEN

To evaluate the concept that in vivo transfer of the human carboxylesterase gene will confer sensitivity of a solid tumor to the prodrug CPT-11 (irinotecan), we constructed an adenovirus vector (AdCMV.CE) carrying the human carboxylesterase gene driven by the cytomegalovirus (CMV) promoter, infected A549 human lung adenocarcinoma cells in vitro and in vivo, and evaluated cell growth over time. AdCMV.CE produced a functional carboxylesterase protein in A549 cells in vitro and in vivo as evidenced by ability of lysates from the infected cells to convert CPT-11 to its active metabolite SN-38. The AdCMV.CE vector effectively suppressed A549 cell growth in vitro in the presence of CPT-11. Cell mixing studies demonstrated that when as few as 10% of cells expressed the human carboxylesterase gene, there was bystander growth suppression in the presence of CPT-11. Consistent with these in vitro observations, when AdCMV.CE was directly injected into established subcutaneous A549 tumors in nude mice receiving CPT-11, there was 35% reduction in tumor size at day 27 compared to controls, and a 41% reduction at day 34 (P < 0.01, both comparisons to controls). Similar observations were made with the cell line H157 and HeLa. These observations suggest that local gene transfer of the human carboxylesterase gene and concomitant local administration of CPT-11 may have potential as a strategy for control of the growth of solid tumors.


Asunto(s)
Antineoplásicos Fitogénicos/metabolismo , Antineoplásicos Fitogénicos/uso terapéutico , Camptotecina/análogos & derivados , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Terapia Genética , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Profármacos/metabolismo , Profármacos/uso terapéutico , Adenoviridae/genética , Animales , Camptotecina/metabolismo , Camptotecina/uso terapéutico , Carboxilesterasa , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Irinotecán , Ratones , Ratones Desnudos , Neoplasias/metabolismo , Células Tumorales Cultivadas
6.
J Clin Invest ; 103(4): 579-87, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10021467

RESUMEN

Primary fibroblasts are not efficiently transduced by subgroup C adenovirus (Ad) vectors because they express low levels of the high-affinity Coxsackie virus and adenovirus receptor (CAR). In the present study, we have used primary human dermal fibroblasts as a model to explore strategies by which Ad vectors can be designed to enter cells deficient in CAR. Using an Ad vector expressing the human CAR cDNA (AdCAR) at high multiplicity of infection, primary fibroblasts were converted from being CAR deficient to CAR sufficient. Efficiency of subsequent gene transfer by standard Ad5-based vectors and Ad5-based vectors with alterations in penton and fiber was evaluated. Marked enhancement of binding and transgene expression by standard Ad5 vectors was achieved in CAR-sufficient fibroblasts. Expression by AdDeltaRGDbetagal, an Ad5-based vector lacking the arginine-glycine-aspartate (RGD) alphaV integrin recognition site from its penton base, was achieved in CAR-sufficient, but not CAR-deficient, cells. Fiber-altered Ad5-based vectors, including (a) AdF(pK7)betagal (bearing seven lysines on the end of fiber) (b) AdF(RGD)betagal (bearing a high-affinity RGD sequence on the end of fiber), and (c) AdF9sK betagal (bearing a short fiber and Ad9 knob), demonstrated enhanced gene transfer in CAR-deficient fibroblasts, with no further enhancement in CAR-sufficient fibroblasts. Together, these observations demonstrate that CAR deficiency on Ad targets can be circumvented either by supplying CAR or by modifying the Ad fiber to bind to other cell-surface receptors.


Asunto(s)
Adenovirus Humanos , Proteínas de la Cápside , Técnicas de Transferencia de Gen , Vectores Genéticos , Receptores Virales/metabolismo , Adenovirus Humanos/metabolismo , Cápside/genética , Cápside/metabolismo , Células Cultivadas , Fibroblastos/citología , Expresión Génica , Humanos , Receptores Virales/genética , Transgenes , Regulación hacia Arriba
7.
J Clin Invest ; 104(9): 1245-55, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10545523

RESUMEN

We sought to evaluate the ability of an E1(-), E3(-) adenovirus (Ad) vector (Ad(GV)CFTR.10) to transfer the normal human cystic fibrosis transmembrane conductance regulator (CFTR) cDNA to the airway epithelium of individuals with cystic fibrosis (CF). We administered Ad(GV)CFTR.10 at doses of 3 x 10(6) to 2 x 10(9) plaque-forming units over 9 months by endobronchial spray to 7 pairs of individuals with CF. Each 3-month cycle, we measured vector-derived versus endogenous CFTR mRNA in airway epithelial cells prior to therapy, as well as 3 and 30 days after therapy. The data demonstrate that (a) this strategy appears to be safe; (b) after the first administration, vector-derived CFTR cDNA expression in the CF airway epithelium is dose-dependent, with greater than 5% endogenous CFTR mRNA levels at the higher vector doses; (c) expression is transient, lasting less than 30 days; (d) expression can be achieved with a second administration, but only at intermediate doses, and no expression is observed with the third administration; and (e) the progressive lack of expression with repetitive administration does not closely correlate with induction of systemic anti-Ad neutralizing antibodies. The major advantage of an Ad vector is that it can deliver sufficient levels of CFTR cDNA to the airway epithelium so that CFTR expression protects the lungs from the respiratory manifestations of CF. However, this impressive level of expression is linked to the challenging fact that expression is limited in time. Although this can be initially overcome by repetitive administration, unknown mechanisms eventually limit this strategy, and further repetitive administration does not lead to repetitive expression.


Asunto(s)
Bronquios/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Fibrosis Quística/terapia , Terapia Genética/métodos , Tráquea/metabolismo , Adenoviridae/genética , Adolescente , Adulto , Estudios de Cohortes , Regulador de Conductancia de Transmembrana de Fibrosis Quística/administración & dosificación , Regulador de Conductancia de Transmembrana de Fibrosis Quística/inmunología , ADN Complementario/genética , Relación Dosis-Respuesta a Droga , Epitelio/metabolismo , Femenino , Vectores Genéticos/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Modelos Genéticos , Reacción en Cadena de la Polimerasa , ARN Mensajero/metabolismo , Recombinación Genética , Factores de Tiempo
8.
AJNR Am J Neuroradiol ; 28(7): 1232-6, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17698521

RESUMEN

BACKGROUND AND PURPOSE: Late infantile neuronal ceroid lipofuscinosis (LINCL), a form of Batten disease, is a fatal neurodegenerative genetic disorder, diagnosed via DNA testing, that affects approximately 200 children in the United States at any one time. This study was conducted to evaluate whether quantitative data derived by diffusion-weighted MR imaging (DWI) techniques can supplement clinical disability scale information to provide a quantitative estimate of neurodegeneration, as well as disease progression and severity. MATERIALS AND METHODS: This study prospectively analyzed 32 DWI examinations from 18 patients having confirmed LINCL at various stages of disease. A whole-brain apparent diffusion coefficient (ADC) histogram was fitted with a dual Gaussian function combined with a function designed to model voxels containing a partial volume fraction of brain parenchyma versus CSF. Previously published whole-brain ADC values of age-matched control subjects were compared with those of the LINCL patients. Correlations were tested between the peak ADC of the fitted histogram and patient age, disease severity, and a CNS disability scale adapted for LINCL. RESULTS: ADC values assigned to brain parenchyma were higher than published ADC values for age-matched control subjects. ADC values between patients and control subjects began to differ at 5 years of age based on 95% confidence intervals. ADC values had a nearly equal correlation with patient age (R2=0.71) and disease duration (R2=0.68), whereas the correlation with the central nervous system disability scale (R2=0.27) was much weaker. CONCLUSION: This study indicates that brain ADC values acquired using DWI may be used as an independent measure of disease severity and duration in LINCL.


Asunto(s)
Encéfalo/patología , Imagen de Difusión por Resonancia Magnética/métodos , Interpretación de Imagen Asistida por Computador/métodos , Lipofuscinosis Ceroideas Neuronales/diagnóstico , Índice de Severidad de la Enfermedad , Adolescente , Niño , Preescolar , Femenino , Humanos , Masculino , Lipofuscinosis Ceroideas Neuronales/clasificación , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
9.
Circ Res ; 88(9): 903-10, 2001 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-11348999

RESUMEN

Intravascular introduction of replication-deficient adenoviral vectors (Advectors) provides an ideal model of delivery of transgenes for the treatment of various vascular abnormalities. On the basis of the knowledge that Advectors can induce inflammatory responses after intravascular administration, we speculated that cellular activation by Advector infection could directly modulate the endothelial cell (EC) adhesion molecule/chemokine expression repertoire. Infection of human umbilical vein ECs or bone marrow microvascular ECs with an E1(-)E4(+) Advector resulted in the upregulation of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and CD34, but not E-selectin, P-selectin, CD36, CD13, CD44, HLA-DR or PECAM. Upregulation of ICAM-1, VCAM-1, and CD34 was apparent 12 hours after infection and persisted for weeks after infection. Selective induction of adhesion molecules was mediated by the presence of the E4 gene in the Advector, because infection of ECs with an E1(-)E4(-) Advector had no effect on adhesion molecule expression. ECs infected with E1(-)E4(+) Advector, but not those infected with E1(-)E4(-) Advector, supported the adhesion of leukocytes. Monoclonal antibodies to ICAM-1 and VCAM-1 inhibited adhesion of leukocytes to E1(-)E4(+)-infected ECS: Infection of the ECs with E1(-)E4(+) Advector, but not E1(-)E4(-) Advector, resulted in downregulation of expression of chemocytokines, including interleukin-8, MCP-1, RANTES, and GM-CSF. Nonetheless, a large number of leukocytes migrated through ECs infected with E1(-)E4(+), but not those infected with E1(-)E4(l-), in response to exogenous chemokines. These results demonstrate that infection of ECs with E1(-)E4(+) Advectors, but not E1(-)E4(-) Advectors, may directly augment inflammatory responses by upregulating expression of adhesion molecules and enhancing migration through Advector-infected ECs and suggest that E1(-)E4(-) Advectors may be a better choice for gene-transfer strategies directed to the ECS:


Asunto(s)
Proteínas E1 de Adenovirus/genética , Proteínas E4 de Adenovirus/genética , Endotelio Vascular/metabolismo , Leucocitos/metabolismo , Proteínas/metabolismo , Adenoviridae/genética , Antígenos CD34/genética , Antígenos CD34/metabolismo , Adhesión Celular , Movimiento Celular , Células Cultivadas , Quimiocinas/genética , Quimiocinas/metabolismo , Regulación hacia Abajo , Endotelio Vascular/citología , Regulación de la Expresión Génica , Vectores Genéticos/genética , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos/citología , Proteínas/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Factores de Tiempo , Transfección , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
10.
Cancer Res ; 58(19): 4368-74, 1998 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-9766666

RESUMEN

To evaluate the concept that transfer of the human carboxylesterase (CE) gene will overcome the drug resistance of a solid tumor to CPT-11 (irinotecan), we used an adenovirus vector (AdCMV.CE) carrying human CE cDNA to infect CPT-11-resistant A549 human adenocarcinoma cells (A549/CPT) in vitro and in vivo and evaluated cell growth over time. The A549/CPT cells, selected by stepwise and continuous exposure of parental A549 cells to CPT-11 over 10 months, had a 6-fold resistance to CPT-11 and 42% CE activity in comparison with parental A549 cells. AdCMV.CE infection resulted in an increase in functional CE protein in resistant cells in vitro that was sufficient to convert CPT-11 to its active metabolite, SN-38, and effectively suppressed resistant cell growth in vitro in the presence of CPT-11. When AdCMV.CE was directly injected into established s.c. resistant A549-based tumors in nude mice receiving CPT-11, there was a 1.8-fold reduction in tumor size at day 20 compared to that of controls (P < 0.05). These observations suggest that adenovirus-mediated gene transfer of the human CE gene and concomitant administration of CPT-11 may have potential as a strategy for local control of acquired CPT-11 resistance of solid tumors.


Asunto(s)
Antineoplásicos Fitogénicos/toxicidad , Antineoplásicos/toxicidad , Camptotecina/análogos & derivados , Hidrolasas de Éster Carboxílico/metabolismo , Adenocarcinoma , Adenovirus Humanos , Antineoplásicos Fitogénicos/farmacocinética , Camptotecina/farmacocinética , Camptotecina/toxicidad , Carboxilesterasa , Hidrolasas de Éster Carboxílico/genética , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Clonales , ADN-Topoisomerasas de Tipo I/metabolismo , ADN Complementario , Resistencia a Antineoplásicos , Vectores Genéticos , Humanos , Irinotecán , Neoplasias Pulmonares , Proteínas Recombinantes/metabolismo , Transfección/métodos , Células Tumorales Cultivadas
11.
Biochim Biophys Acta ; 725(1): 168-77, 1983 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-6313050

RESUMEN

Escherichia coli grown anaerobically with trimethylamine N-oxide (TMAO) as a terminal electron acceptor develops a new cytochrome pathway in addition to the aerobic respiratory pathways which are still formed. Formate, NADH, and possibly other substrates derived from glucose, supply electrons to this pathway. Cytochromes with alpha-absorption peaks at about 548, 552, 554 and 557 nm are rapidly reoxidized by TMAO in a reaction which is not inhibited by 2-n-heptyl -4-hydroxyquinone N-oxide. CuSO4 inhibits the reoxidation by TMAO of the first two of these cytochromes. This suggests that the pathway of electron transfer leading to the reduction of TMAO is: substrates leads to cytochromes 548,552 leads to cytochromes 554,557 leads to trimethylamine-N-oxide reductase leads to TMAO. These cytochromes, but not those of the aerobic respiratory pathways, are reoxidized by the membrane-impermeant oxidant ammonium persulfate in intact cells. This suggests that the cytochromes of the TMAO reduction pathway and/or trimethylamine-N-oxide reductase are situated at the periplasmic surface of the cytoplasmic membrane of E. coli.


Asunto(s)
Citocromos/metabolismo , Escherichia coli/metabolismo , Metilaminas/farmacología , Anaerobiosis , Grupo Citocromo b/metabolismo , Grupo Citocromo c/metabolismo , Ditionita , Transporte de Electrón/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Cinética , Oxidación-Reducción
12.
Circulation ; 100(5): 468-74, 1999 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-10430759

RESUMEN

BACKGROUND: Therapeutic angiogenesis, a new experimental strategy for the treatment of vascular insufficiency, uses the administration of mediators known to induce vascular development in embryogenesis to induce neovascularization of ischemic adult tissues. This report summarizes a phase I clinical experience with a gene-therapy strategy that used an E1(-)E3(-) adenovirus (Ad) gene-transfer vector expressing human vascular endothelial growth factor (VEGF) 121 cDNA (Ad(GV)VEGF121.10) to induce therapeutic angiogenesis in the myocardium of individuals with clinically significant coronary artery disease. METHODS AND RESULTS: Ad(GV)VEGF121.10 was administered to 21 individuals by direct myocardial injection into an area of reversible ischemia either as an adjunct to conventional coronary artery bypass grafting (group A, n=15) or as sole therapy via a minithoracotomy (group B, n=6). There was no evidence of systemic or cardiac-related adverse events related to vector administration. In both groups, coronary angiography and stress sestamibi scan assessment of wall motion 30 days after therapy suggested improvement in the area of vector administration. All patients reported improvement in angina class after therapy. In group B, in which gene transfer was the only therapy, treadmill exercise assessment suggested improvement in most individuals. CONCLUSIONS: The data are consistent with the concept that direct myocardial administration of Ad(GV)VEGF121.10 to individuals with clinically significant coronary artery disease appears to be well tolerated, and initiation of phase II evaluation of this therapy is warranted.


Asunto(s)
Adenoviridae , Circulación Coronaria , Enfermedad Coronaria/terapia , Factores de Crecimiento Endotelial/genética , Terapia Genética/métodos , Vectores Genéticos , Linfocinas/genética , Neovascularización Fisiológica/genética , Adulto , Anciano , Anciano de 80 o más Años , Terapia Combinada , Puente de Arteria Coronaria , Enfermedad Coronaria/fisiopatología , Enfermedad Coronaria/cirugía , ADN Complementario/biosíntesis , Prueba de Esfuerzo , Femenino , Humanos , Inyecciones , Masculino , Persona de Mediana Edad , Miocardio , Índice de Severidad de la Enfermedad , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
13.
Hum Gene Ther ; 12(10): 1251-63, 2001 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-11440619

RESUMEN

We have shown that dendritic cells (DCs) genetically engineered with a recombinant adenovirus vector (Ad) to express CD40 ligand (CD40L) elicit specific humoral immunity against the Pseudomonas aeruginosa laboratory strain PAO1, without CD4(+) T cell help. In the present study, using several different strains of P. aeruginosa, we examine whether this strategy is generally applicable to enhancing clinically relevant pathogen-specific immunity. Mice immunized with DCs modified with CD40L and pulsed with heat-killed P. aeruginosa clinical strain PA514, originally isolated from the sputum of an individual with cystic fibrosis, survived lethal respiratory challenge with PA514-impregnated agar beads. Consistent with this effective in vivo protection, the immunized mice generated high levels of serum isotype-switched antibodies directed against PA514 without concomitant nonspecific elevations of total serum immunoglobulin levels. The CD40L genetically engineered DCs pulsed with seven of eight different strains of P. aeruginosa afforded significant, albeit variable, cross-protection against lethal respiratory challenge with a clinical (PA514) or laboratory (PAO1) strain of P. aeruginosa. CD40L genetically modified DCs pulsed with a clinical (PA514) or laboratory (PAO1) strain of P. aeruginosa initiated cross-reacting antibody responses against each other, but not against Escherichia coli and vice versa. These observations may be useful in developing vaccines for infectious diseases, including P. aeruginosa infection.


Asunto(s)
Ligando de CD40/biosíntesis , Ligando de CD40/genética , Células Dendríticas/metabolismo , Pseudomonas aeruginosa/metabolismo , Vacunas de ADN/genética , Adenoviridae/genética , Animales , Células de la Médula Ósea/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Femenino , Humanos , Inmunoglobulinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Infecciones por Pseudomonas/prevención & control , Pseudomonas aeruginosa/inmunología , Factores de Tiempo
14.
Hum Gene Ther ; 12(17): 2035-49, 2001 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-11747595

RESUMEN

Although tumor necrosis factor alpha (TNF-alpha) is a potent cytokine with a myriad of innate immune antitumor properties, systemic administration of TNF-alpha is associated with significant toxicity, limiting the use of the TNF-alpha protein as an antitumor therapeutic. On the basis of the knowledge that dendritic cells (DCs) play a central role in initiating antitumor adaptive immune responses, we hypothesized that intratumoral administration of low doses of an adenovirus encoding TNF-alpha (AdTNF-alpha) together with syngeneic DCs would act synergistically to suppress preexisting tumors. As a model, four different tumor cell lines, all resistant in vitro to the TNF-alpha protein, were implanted in syngeneic mice, and established tumors received intratumor AdTNF-alpha alone or in combination with DCs. At high doses (10(9) PFU), AdTNF-alpha alone suppressed tumor growth, but was associated with systemic toxicity. A 100-fold lower AdTNF-alpha concentration (10(7) PFU) or high doses of the control vector AdNull had no systemic toxicity, but also minimal suppression of tumor growth. In contrast, local administration of the low dose (10(7) PFU) of AdTNF-alpha in combination with syngeneic DCs (AdTNF-alpha + DCs) elicited marked tumor suppression without toxicity. Administration of AdTNF-alpha + DCs into tumors elicited tumor-specific cytotoxic T cells and protected animals against subsequent challenge with the same tumor, suggesting that AdTNF-alpha + DC therapy induced tumor-specific adaptive immune host responses. Consistent with this concept, studies with syngeneic knockout mice showed that MHC class I molecules on DCs as well as CD8(+) T cells were necessary for the antitumor effect of intratumor AdTNF-alpha + DCs. These data demonstrate that the combination of intratumoral administration of the TNF-alpha cDNA together with naive DCs can evoke tumor suppression without systemic toxicity, providing a new paradigm for the use of TNF-alpha as antitumor therapy.


Asunto(s)
Adenoviridae/genética , Células Dendríticas/inmunología , Vectores Genéticos/administración & dosificación , Neoplasias/inmunología , Neoplasias/terapia , Factor de Necrosis Tumoral alfa/efectos adversos , Factor de Necrosis Tumoral alfa/uso terapéutico , Animales , División Celular , Supervivencia Celular , Células Dendríticas/trasplante , Femenino , Terapia Genética/métodos , Inmunoterapia Adoptiva/métodos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Fluorescente , Trasplante de Neoplasias , Neoplasias/genética , Neoplasias/patología , Linfocitos T Citotóxicos/inmunología , Factores de Tiempo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor de Necrosis Tumoral alfa/genética
15.
Hum Gene Ther ; 9(3): 367-78, 1998 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-9508054

RESUMEN

The pathogenic agent, adenovirus (Ad), has taken on a new role as a vector for gene transfer in both laboratory and clinical settings. To help understand the intracellular pathways and fate of Ad gene transfer vectors, we covalently conjugated fluorophores to E1-, E3- Ad vectors and used quantitative fluorescence microscopy to assess essential steps of Ad vector gene transfer to the A549 human epithelial lung cell line including binding, internalization, escape from endosomes, translocation to the nucleus, dissociation of capsids and gene expression. The data demonstrate that Ad internalizes with a t1/2 2.5 min, breaks out of endosomes early, likely prior to endosome-endosome fusion, exhibits sustained, intracellular velocities averaging 0.58 microm/sec, and translocates to the nucleus with >80% of internalized fluorophore demonstrating nuclear localization within 60 min of infection. Interestingly, 24 hr after infection, half of the initially internalized fluorescence was detected but lacked nuclear localization, suggesting that the capsid is released from the nucleus and is likely degraded. Fluorescent labeling of virions provides a novel quantitative, morphological strategy to characterize the interaction of gene transfer vectors with the intracellular environment.


Asunto(s)
Adenoviridae/fisiología , Carbocianinas/metabolismo , Colorantes Fluorescentes/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/fisiología , Adenoviridae/metabolismo , Núcleo Celular/metabolismo , Citosol/metabolismo , Endosomas/metabolismo , Vectores Genéticos/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Factores de Tiempo , Células Tumorales Cultivadas , Virión
16.
Hum Gene Ther ; 9(6): 823-33, 1998 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-9581905

RESUMEN

Vascular endothelial growth factor (VEGF), a potent angiogenic mediator, is overexpressed in most solid tumors. On the basis of the knowledge that solid tumor growth beyond a small volume is critically dependent on angiogenesis, and that adenovirus (Ad) vectors can mediate efficient in vivo gene transfer and expression, we hypothesized that Ad-mediated transfer of a secreted form of the extracellular domain of the flt-1 VEGF receptor (Adsflt) would suppress tumor growth on a regional basis. To evaluate this concept, three tumor models were examined using a murine colon carcinoma cell line and syngeneic BALB/c mice. First, mice with preestablished splenic CT26.CL25 tumors and liver metastases were given Adsflt on AdNull intravenously and, after 15 days, spleens and livers were harvested to quantify tumor burden. Adslft-treated animals had minimal residual splenic tumors and liver metastases; in contrast, control animals had bulky splenic tumors and extensive liver metastases (p < 0.003). Second, mice with preestablished lung metastases showed a significant reduction in pulmonary metastases with regionally administered Adslft (intratracheal, p < 0.02) but not when the vector was systemically administered (intravenous, p > 0.9). Finally, mice with primary subcutaneous tumors treated with intratumoral administration of Adslft showed significant tumor suppression (p < 0.05) not observed in AdNull-treated mice or mice given Adslft intravenously (p > 0.3). We conclude that Ad-mediated in vivo regional delivery of a secreted form of the extracellular domain of the flt-1 VEGF receptor can effectively inhibit regional tumor growth, a strategy that may provide a means to control tumor growth within the treated organ without the risk of systemic antiangiogenesis.


Asunto(s)
Terapia Genética , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica/prevención & control , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Adenoviridae , Animales , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , ADN Complementario/administración & dosificación , Técnicas de Transferencia de Gen , Vectores Genéticos , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/secundario , Neoplasias Cutáneas/terapia , Células Tumorales Cultivadas , Receptor 1 de Factores de Crecimiento Endotelial Vascular
17.
Hum Gene Ther ; 8(1): 99-109, 1997 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-8989999

RESUMEN

Effective gene transfer and expression following repetitive administration of adenoviral (Ad) vectors in experimental animals is limited by anti-Ad neutralizing antibodies. Knowing that anti-Ad humoral immunity is serotype-specific, we hypothesized that anti-Ad neutralizing immunity could be circumvented using Ad vectors of different serotypes (Ad2, Ad5) within the same subgroup (C) to transfer and express beta-glucuronidase (beta glu) in the lung. Sprague-Dawley rats received an intratracheal administration of either Ad2 beta glu or Ad5 beta glu, and, 14 days later, repeat administration of either the same vector or a vector of a different serotype. Analysis of serum and bronchoalveolar lavage fluid following initial vector administration demonstrated systemic and local serotype-specific neutralizing antibodies. For both the Ad2 and Ad5 vectors, beta glu expression 24 hr following the second administration of the same serotype was < 30% of that of naive animals. In contrast, beta glu expression 24 hr following second administration of a different serotype Ad vector was similar to expression at 24 hr of naive animals receiving a single administration (Ad5 beta glu followed by Ad2 beta glu, as well as Ad2 beta glu followed by Ad5 beta glu; p > 0.2 both comparisons). Although the alternative serotype bypassed anti-Ad neutralizing immunity, persistence of expression was reduced compared to that following administration to naive animals. Compatible with this observation, systemic administration of the same vectors to C57B1/6 mice demonstrated induction of cytotoxic T lymphocytes directed against the beta glu transgene, as well as products of the Ad genome. Interestingly, intratracheal administration of vectors with different serotypes and different transgenes to rats resulted in longer expression (but still not normalized) compared to that achieved with vectors of different serotypes but the same transgene. These observations demonstrate that alternate use of Ad vectors from different serotypes within the same subgroup can circumvent anti-Ad humoral immunity to permit effective gene transfer after repeat administration, although the chronicity of expression is limited, likely by cellular immune process directed against both the transgene and viral gene products expressed by the vector.


Asunto(s)
Adenoviridae/inmunología , Técnicas de Transferencia de Gen , Inmunidad/inmunología , Adenoviridae/genética , Animales , Anticuerpos/sangre , Anticuerpos/farmacología , Líquido del Lavado Bronquioalveolar/química , Células Cultivadas , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Regulación Viral de la Expresión Génica/genética , Vectores Genéticos/genética , Glucuronidasa/genética , Glucuronidasa/metabolismo , Pulmón/virología , Ratones , Ratones Endogámicos , Ratas , Ratas Sprague-Dawley , Serotipificación , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Proteínas Virales/inmunología , Proteínas Virales/metabolismo
18.
Hum Gene Ther ; 10(8): 1331-48, 1999 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-10365664

RESUMEN

A gene therapy strategy involving direct myocardial administration of an adenovirus (Ad) vector encoding the vascular endothelial growth factor 121 cDNA (Ad(GV)VEGF121.10) has been shown to be capable of "biological revascularization" of ischemic myocardium in an established porcine model [Mack, C.A. (1998). J. Thorac. Cardiovasc. Surg. 115, 168-177]. The present study evaluates the local and systemic safety of this therapy in this porcine ischemia model and in normal mice. Myocardial ischemia was induced in Yorkshire swine with an ameroid constrictor 21 days prior to vector administration. Ad(GV)VEGF121.10 (10(9) or 10(10) PFU), Ad5 wild type (10(9) PFU), AdNull (control vector with no transgene; 10(9) PFU), saline, or no injection (naive) was administered in 10 sites in the ischemic, circumflex distribution of the myocardium. Toxicity was assessed by survival, serial echocardiography, blood analyses, and myocardial and liver histology at 3 and 28 days after vector administration. All pigs survived to sacrifice, except for one animal in the Ad(GV)VEGF121.10 (10(10) PFU) group, which died as a result of oversedation. Echocardiograms of Ad(GV)VEGF121.10-treated pigs demonstrated no differences in pericardial effusion, mitral valve regurgitation, or regional wall motion compared with control pigs. Intramyocardial administration of Ad(GV)VEGF121.10 included only minimal myocardial inflammation and necrosis, and no hepatic inflammation or necrosis. Only a mild elevation of the white blood cell count was encountered on day 3, which was transient and self-limited in the Ad(GV)VEGF121.10 group as compared with the saline-treated animals. As a measure of inadvertent intravascular administration of vector, normal C57/BL6 mice received intravenous Ad(GV)VEGF121.10 (10(4), 10(6), 5 x 10(7), or 10(9) PFU), AdNull (5 x 10(7) or 10(9) PFU), or saline. Toxicity was assessed by survival, blood analyses, and organ histology at 3 and 7 days after vector administration. A separate group of C57/BL6 mice received intravenous AdmVEGF164 (Ad vector encoding the murine VEGF164 cDNA), Ad(GV)VEGF121.10, AdNull (10(8) PFU each group), or saline to assess duration of expression and safety of a homologous transgene. All mice survived to sacrifice except for 40% of the mice in the highest (10(9) PFU; a dose more than 10(3)-fold higher by body weight than the efficacious dose in pigs) Ad(GV)VEGF121.10 dose group, which died on days 5-6 after vector administration. The only differences seen in the blood analyses between treated and control mice were in the very high Ad(GV)VEGF121.10 dose group (10(9) PFU), which demonstrated an anemia as well as an increase in alkaline phosphatase when compared with all other treatment groups. Hepatic VEGF levels by ELISA in AdmVEGF164-treated mice did not persist beyond 14 days after vector administration, suggesting that persistent expression of a homologous VEGF gene transferred with an Ad vector is not a significant safety risk. Although this is not a chronic toxicity study, these data demonstrate the safety of direct myocardial administration of Ad(GV)VEGF121.10, and support the potential use of this strategy to treat human myocardial ischemia.


Asunto(s)
Adenovirus Humanos , Factores de Crecimiento Endotelial/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Linfocinas/genética , Isquemia Miocárdica/terapia , Animales , Seguridad de Productos para el Consumidor , Modelos Animales de Enfermedad , Ecocardiografía , Pruebas Hematológicas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Isquemia Miocárdica/diagnóstico por imagen , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Miocardio/patología , Porcinos , Transgenes , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
19.
J Invest Dermatol ; 113(6): 999-1005, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10594743

RESUMEN

Dendritic antigen-presenting cells derived from epidermis (Langerhans cells), bone marrow, and peripheral blood can present a wide variety of antigens, including tumor-associated antigens, for various immune responses. The development and function of dendritic cells is dependent upon a number of cytokines including granulocyte-macrophage-colony-stimulating factor. For example, Langerhans cells can present tumor-associated antigens for the induction of substantial in vivo anti-tumor immunity but only after activation in vitro by granulocyte-macrophage-colony-stimulating factor. Thus, we reasoned that insertion of a cDNA for granulocyte-macrophage-colony-stimulating factor into dendritic antigen-presenting cells may allow for autocrine stimulation and increased antigen-presenting capability. To test this possibility, we utilized an adenovirus vector to insert a cDNA for murine granulocyte-macrophage-colony-stimulating factor into the dendritic cell lines XS52-4D and XS106 (derived from neonatal mouse epidermis), bone marrow-derived dendritic cells, and epidermal cells that contain Langerhans cells. Infection of each of these cell types resulted in release of abundant quantities of granulocyte-macrophage-colony-stimulating factor. XS52-4D and XS106 cells infected with adenovirus granulocyte-macrophage-colony-stimulating factor exhibited prolonged dendrites and greater expression of major histocompatibility complex class II molecules and CD86 compared with cells infected with a null vector. Granulocyte-macrophage-colony-stimulating factor cDNA-containing XS cells, bone marrow-derived dendritic cells, and epidermal cells had more potent alloantigen presenting capability than cells infected with a null vector. Most importantly, granulocyte-macrophage-colony-stimulating factor gene-transferred epidermal cells were able to present tumor-associated antigens for in vivo anti-tumor immunity against challenge with the S1509a spindle-cell tumor whereas null vector-infected cells were unable to prime for immunity. These results suggest that introduction of a cDNA for granulocyte-macrophage-colony-stimulating factor into dendritic cells may be an effective means to augment their antigen-presenting capability and that granulocyte-macrophage-colony-stimulating factor gene-transfer- red epidermal cells may be useful in tumor vaccination strategies.


Asunto(s)
Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Células Dendríticas/fisiología , Epidermis/inmunología , Técnicas de Transferencia de Gen , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Neoplasias Experimentales/inmunología , Animales , Femenino , Inmunización , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL
20.
Arch Neurol ; 58(11): 1793-8, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11708986

RESUMEN

Late infantile neuronal ceroid lipofuscinosis is a progressive childhood neurodegenerative disorder characterized by intracellular accumulation of autofluorescent material resembling lipofuscin in neuronal cells. This report summarizes the new therapies under consideration for late infantile neuronal ceroid lipofuscinosis, with a focus on strategies for in vivo gene therapy for the retinal and central nervous system manifestations of the disease.


Asunto(s)
Endopeptidasas/genética , Terapia Genética , Lipofuscinosis Ceroideas Neuronales/genética , Lipofuscinosis Ceroideas Neuronales/terapia , Adulto , Aminopeptidasas , Animales , Niño , Preescolar , Ensayos Clínicos como Asunto , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas , Endopeptidasas/uso terapéutico , Vectores Genéticos , Humanos , Serina Proteasas , Trasplante de Células Madre , Tripeptidil Peptidasa 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA