Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Appl Microbiol ; 123(1): 66-79, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28245091

RESUMEN

AIM: To evaluate an antibiotic inactivation strategy to protect the gut microbiome from antibiotic-mediated damage. METHODS AND RESULTS: SYN-004 (ribaxamase) is an orally delivered beta-lactamase intended to degrade penicillins and cephalosporins within the gastrointestinal tract to protect the microbiome. Pigs (20 kg, n = 10) were treated with ceftriaxone (CRO) (IV, 50 mg kg-1 , SID) for 7 days and a cohort (n = 5) received ribaxamase (PO, 75 mg, QID) for 9 days beginning the day before antibiotic administration. Ceftriaxone serum levels were not statistically different in the antibiotic-alone and antibiotic + ribaxamase groups, indicating ribaxamase did not alter systemic antibiotic levels. Whole-genome metagenomic analyses of pig faecal DNA revealed that CRO caused significant changes to the gut microbiome and an increased frequency of antibiotic resistance genes. With ribaxamase, the gut microbiomes were not significantly different from pretreatment and antibiotic resistance gene frequency was not increased. CONCLUSION: Ribaxamase mitigated CRO-mediated gut microbiome dysbiosis and attenuated propagation of the antibiotic resistance genes in pigs. SIGNIFICANCE AND IMPACT OF THE STUDY: Damage of the microbiome can lead to overgrowth of pathogenic organisms and antibiotic exposure can promote selection for antibiotic-resistant micro-organisms. Ribaxamase has the potential to become the first therapy designed to protect the gut microbiome from antibiotic-mediated dysbiosis and reduce emergence of antibiotic resistance.

2.
Nat Genet ; 5(4): 397-402, 1993 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-8298650

RESUMEN

Gene therapy strategies designed to combat haemophilia B, caused by defects in clotting factor IX, have so far concentrated on ex vivo approaches. We have now evaluated adenoviral vector-mediated expression of human factor IX in vivo. Injection of the vector Av1H9B, which encodes human factor IX cDNA, into the tail veins of mice resulted in efficient liver transduction and plasma levels of human factor IX that would be therapeutic for haemophilia B patients. However, levels slowly declined to baseline by nine weeks and were not re-established by a second vector injection. These results address both the advantages and obstacles to the use of adenoviral vectors for treatment of haemophilia B.


Asunto(s)
Adenovirus Humanos/genética , Factor IX/metabolismo , Vectores Genéticos , Animales , Southern Blotting , Ensayo de Inmunoadsorción Enzimática , Factor IX/genética , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Ratones , Ratones Endogámicos C57BL
3.
Mol Cell Biol ; 10(2): 464-73, 1990 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-2153917

RESUMEN

The human insulinlike growth factor I receptor was overexpressed in NIH 3T3 cells as well as human and rat primary fibroblast strains. The NIH 3T3 cells displayed a ligand-dependent, highly transformed phenotype. When exposed to insulinlike growth factor I or supraphysiologic levels of insulin, NIH 3T3 cells that expressed high levels of receptors formed aggregates in tissue culture dishes, colonies in soft agar, and tumors in nude mice. Expression of 1 million receptors per cell, a 40-fold increase above the base-line level, was required for anchorage-independent growth. Primary fibroblasts that expressed high levels of receptors displayed a ligand-dependent change in morphology and an increase in saturation density but did not acquire a fully transformed phenotype. The results demonstrate that when amplified, this ubiquitous growth factor receptor behaves like an oncogenic protein and is capable of promoting neoplastic growth in vivo.


Asunto(s)
Transformación Celular Neoplásica , Expresión Génica , Genes , Factor I del Crecimiento Similar a la Insulina/metabolismo , Receptores de Superficie Celular/genética , Somatomedinas/metabolismo , Animales , División Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Vectores Genéticos , Humanos , Insulina/farmacología , Factor I del Crecimiento Similar a la Insulina/farmacología , Cinética , Ligandos , Ratones , Ratones Endogámicos , Fenotipo , Ratas , Receptores de Superficie Celular/metabolismo , Receptores de Somatomedina , Transfección
4.
Oncogene ; 7(9): 1859-66, 1992 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-1354348

RESUMEN

Amplification of the HER-2 (c-erbB-2) gene and overexpression of the p185HER-2 gene product is found in approximately one-third of primary human breast and ovarian cancers and is associated with a poor clinical outcome of early relapse and death. The HER-2 gene encodes a cell-surface growth factor receptor with intrinsic tyrosine kinase activity. Wild-type human HER-2 has been shown to act as a potent oncogene when over-expressed in mouse fibroblasts. Recent data suggest that the mechanism by which HER-2 mediates transformation requires the interaction of the epidermal growth factor (EGF) receptor. To test whether overexpression of normal human HER-2 can transform cells independently of the EGF receptor, we have introduced multiple copies of HER-2 into the EGF receptor-negative cell line, NR6, and have performed assays for both transformation and tumorigenicity. Engineered NR6 cells that overexpress the HER-2 gene product display a highly transformed and tumorigenic phenotype as compared with control cells. Additionally, a monoclonal antibody to the extracellular domain of the HER-2 receptor is able to inhibit the proliferation of the overexpressing cells in vitro as well as tumor growth in vivo. This study provides clear evidence that HER-2-mediated transformation can be achieved independently of the EGF receptor.


Asunto(s)
Transformación Celular Neoplásica , Receptores ErbB/fisiología , Proteínas Proto-Oncogénicas/genética , Proto-Oncogenes , Expresión Génica , Humanos , Receptor ErbB-2 , Células Tumorales Cultivadas
5.
Diabetes ; 50(10): 2287-95, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11574410

RESUMEN

Hyperglycemia of diabetes is caused in part by perturbation of hepatic glucose metabolism. Hepatic glucokinase (GK) is an important regulator of glucose storage and disposal in the liver. GK levels are lowered in patients with maturity-onset diabetes of the young and in some diabetic animal models. Here, we explored the adenoviral vector-mediated overexpression of GK in a diet-induced murine model of type 2 diabetes as a treatment for diabetes. Diabetic mice were treated by intravenous administration with an E1/E2a/E3-deleted adenoviral vector encoding human hepatic GK (Av3hGK). Two weeks posttreatment, the Av3hGK-treated diabetic mice displayed normalized fasting blood glucose levels (95 +/- 4.8 mg/dl; P < 0.001) when compared with Av3Null (135 +/- 5.9 mg/dl), an analogous vector lacking a transgene, and vehicle-treated diabetic mice (134 +/- 8 mg/dl). GK treatment also resulted in lowered insulin levels (632 +/- 399 pg/ml; P < 0.01) compared with the control groups (Av3Null, 1,803 +/- 291 pg/ml; vehicle, 1,861 +/- 392 pg/ml), and the glucose tolerance of the Av3hGK-treated diabetic mice was normalized. No significant increase in plasma or hepatic triglycerides, or plasma free fatty acids was observed in the Av3hGK-treated mice. These data suggest that overexpression of GK may have a therapeutic potential for the treatment of type 2 diabetes.


Asunto(s)
Diabetes Mellitus/genética , Expresión Génica/fisiología , Glucoquinasa/genética , Adenoviridae/genética , Animales , Glucemia/análisis , Diabetes Mellitus/fisiopatología , Ingestión de Alimentos , Ayuno/sangre , Técnicas de Transferencia de Gen , Vectores Genéticos , Glucoquinasa/metabolismo , Glucógeno/metabolismo , Humanos , Insulina/sangre , Hígado/enzimología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Triglicéridos/metabolismo
6.
Diabetes ; 50(8): 1813-20, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11473043

RESUMEN

The enzyme glucokinase (GK) plays a central role in glucose homeostasis. Hepatic GK activity is acutely controlled by the action of the GK regulatory protein (GKRP). In vitro evidence suggests that GKRP reversibly binds to GK and inhibits its activity; however, less is known about the in vivo function of GKRP. To further explore the physiological role of GKRP in vivo, we used an E1/E2a/E3-deficient adenoviral vector containing the cDNA encoding human GKRP (Av3hGKRP). High fat diet-induced diabetic mice were administered Av3hGKRP or a control vector lacking a transgene (Av3Null). Surprisingly, the Av3hGKRP-treated mice showed a significant improvement in glucose tolerance and had lower fasting blood glucose levels than Av3Null-treated mice. A coincident decrease in insulin levels indicated that the Av3hGKRP-treated mice had sharply improved insulin sensitivity. These mice also exhibited lower leptin levels, reduced body weight, and decreased liver GK activity. In vitro experiments indicated that GKRP was able to increase both GK protein and enzymatic activity levels, suggesting that another role for GKRP is to stabilize and/or protect GK. These data are the first to indicate the ability of GKRP to treat type 2 diabetes and therefore have significant implications for future therapies of this disease.


Asunto(s)
Proteínas Portadoras , Diabetes Mellitus Tipo 2/terapia , Terapia Genética , Proteínas/genética , Proteínas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Virus del Sarcoma Aviar/genética , Glucemia/metabolismo , Peso Corporal , Células Cultivadas , Diabetes Mellitus Tipo 2/etiología , Grasas de la Dieta/efectos adversos , Ayuno , Vectores Genéticos , Glucoquinasa/antagonistas & inhibidores , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/terapia , Prueba de Tolerancia a la Glucosa , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Hígado/fisiología , Glucógeno Hepático/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Ratas , Ratas Sprague-Dawley , Virus 40 de los Simios/genética , Transfección , Células Tumorales Cultivadas
7.
Hum Gene Ther ; 2(1): 27-32, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1650585

RESUMEN

The development of liver-directed gene therapy protocols depends upon the ability to transfer genes into a large number of liver cells such that the genes are expressed persistently. We used a retroviral vector to transfer the gene for neomycin phosphotransferase (neo) into mouse liver cells in vivo. Direct injection of the retrovirus preparation into mitotically active (regenerating) liver parenchyma resulted in efficient gene transfer, with neo sequences detectable in the livers of every animal tested 10 weeks to 6 months later. The neo gene was expressed for at least 3 months. This methodology may eventually be applicable to the treatment of human disease.


Asunto(s)
Expresión Génica , Vectores Genéticos , Hígado/metabolismo , Retroviridae/genética , Transfección , Animales , Secuencia de Bases , ADN/análisis , Kanamicina Quinasa , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Fosfotransferasas/biosíntesis , Fosfotransferasas/genética , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Secuencias Repetitivas de Ácidos Nucleicos
8.
Hum Gene Ther ; 7(2): 183-95, 1996 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-8788169

RESUMEN

Hemophilia A results from subnormal levels of blood coagulation factor VIII (FVIII) and is an attractive target for gene therapy. However, progress has been impeded by features of FVIII biology such as low mRNA accumulation and the instability of the protein. We have shown previously that a FVIII adenoviral vector, Av1ALH81, allowed high-level expression of human FVIII in mice sustained for several weeks. Here, we have generated a second FVIII adenoviral vector, Av1ALAPH81, in which an intron was introduced into the FVIII expression cassette. Administration of Av1ALAPH81 to mice resulted in significantly increased FVIII plasma levels, 1,046 +/- 163 ng/ml compared to 307 +/- 93 ng/ml of FVIII detected in mice that received Av1ALH81. Normal FVIII levels in humans are 100-200 ng/ml and therapeutic levels are as low as 10 ng/ml. Therapeutic levels are defined as the amount of FVIII necessary to convert severe hemophilia to a moderate or mild hemophiliac condition. The increased potency of the second FVIII adenoviral vector allowed the administration of significantly lower, less toxic vector doses, while retaining the potential for high FVIII expression. Furthermore, we demonstrate that adenoviral-mediated expression of human FVIII can be limited to the liver by inclusion of a liver-specific promoter, thereby achieving the first step in regulated expression of human FVIII in vivo.


Asunto(s)
Adenovirus Humanos/genética , Factor VIII/genética , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Albúminas/genética , Animales , ADN/genética , Factor VIII/biosíntesis , Factor VIII/metabolismo , Humanos , Intrones/genética , Hígado/metabolismo , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Proteínas Recombinantes de Fusión/biosíntesis
9.
Hum Gene Ther ; 1(1): 31-41, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-2081183

RESUMEN

Three retroviral vectors, containing a human adenosine deaminase (ADA) cDNA linked to either the simian virus 40 (SV40) early promoter, the human cytomegalovirus (CMV) immediate early promoter, or the Moloney murine leukemia virus (MoMLV) promoter, were tested for their ability to express ADA following infection and transplantation of murine bone marrow. Virus was produced by using PA317 amphotropic retrovirus packaging cells. The titer of each of the vectors was similar and no helper virus was detected. Human ADA was expressed in the blood of some animals for 6 months after transplantation of infected marrow, and vector DNA was found in the spleen and in bone marrow from these animals. The percentage of animals expressing human ADA (33%) and the amount of human ADA in blood (1-5% of total ADA) was similar for each of the vectors. These results show that amphotropic vectors are capable of infecting pluripotent hematopoietic stem cells having long-term repopulating ability, and that a variety of promoters allow gene expression following differentiation of these early cells.


Asunto(s)
Adenosina Desaminasa/genética , Trasplante de Médula Ósea , Vectores Genéticos , Retroviridae/genética , Adenosina Desaminasa/biosíntesis , Animales , Secuencia de Bases , Médula Ósea/enzimología , Médula Ósea/microbiología , ADN Viral/análisis , Femenino , Virus Helper/genética , Virus Helper/aislamiento & purificación , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Sondas de Oligonucleótidos , Factores de Tiempo
10.
Hum Gene Ther ; 12(13): 1651-61, 2001 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-11535168

RESUMEN

Hemophilia A patients are typically treated by factor VIII (FVIII) protein replacement, an expensive therapy that induces FVIII-specific inhibitors in approximately 30% of patients with severe hemophilia. FVIII gene therapy has the potential to improve the current treatment protocols. In this report, we used a hemophilia A mouse model to compare the humoral and cellular immune responses between an E1/E2a/E3-deficient adenovirus expressing human FVIII directed by a liver-specific albumin promoter and purified recombinant FVIII protein infusion. Adenovirus-mediated FVIII expression did not elicit detectable CD4+ or CD8+ T cell responses and induced a weak antibody immune response to FVIII. In contrast, FVIII protein administration resulted in a potent anti-FVIII antibody response and moderate CD4+ T cell response. Furthermore, hemophiliac mice preimmunized with FVIII protein infusion to induce anti-FVIII immunity, and subsequently treated by adenovirus-mediated FVIII gene therapy, expressed therapeutic levels of FVIII despite the presence of low levels of anti-FVIII antibodies. No FVIII was detected in the plasma of mice with intermediate or high antibody levels, although anti-FVIII antibody levels in some vector-treated animals declined. The data support the hypothesis that liver-specific gene therapy-mediated expression of FVIII may be less immunogenic than traditional protein replacement therapy.


Asunto(s)
Adenoviridae/genética , Factor VIII/genética , Factor VIII/inmunología , Terapia Genética , Hemofilia A/inmunología , Hemofilia A/terapia , Albúminas/genética , Animales , Anticuerpos/inmunología , Modelos Animales de Enfermedad , Factor VIII/administración & dosificación , Factor VIII/uso terapéutico , Expresión Génica , Vectores Genéticos/genética , Hemofilia A/genética , Humanos , Isotipos de Inmunoglobulinas/biosíntesis , Isotipos de Inmunoglobulinas/inmunología , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , Ovalbúmina/inmunología , Regiones Promotoras Genéticas/genética , Linfocitos T Citotóxicos/inmunología , Factores de Tiempo
11.
Hum Gene Ther ; 10(18): 2999-3011, 1999 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-10609660

RESUMEN

An E1-, E2a-, E3-deleted adenoviral vector (Av3H82) encoding an epitope-tagged B domain-deleted human factor VIII cDNA (flagged FVIII) was evaluated in nonhuman primates. Twelve cynomolgus monkeys received intravenous administration of Av3H82; 6 monkeys received 6 x 10(11) particles/kg and another 6 received 3 x 10(12) particles/kg. Adenoviral vector transduction of the liver was efficient, reproducible, and linearly dose dependent. Physiologic levels of flagged FVIII were readily detected in plasma samples obtained from monkeys that received the higher dose of vector and human FVIII mRNA was detected in their livers. Expression of transgene mRNA was restricted to the liver by the albumin promoter. Although vector DNA was readily detected in the liver of monkeys that received the lower dose, neither human FVIII mRNA nor flagged FVIII protein could be detected. Vector distribution was widespread, with the highest levels observed in liver and spleen. Histopathology, hematology, and serum chemistry analysis identified the liver and blood as major sites of toxicity. Transient mild serum elevations of liver enzymes were observed, along with a dose-dependent inflammatory response in the liver. In addition, mild lymphoid hyperplasia was observed in the spleen. Mild anemia and a transient decrease in platelet count were observed, as was marrow hyperplasia and extramedullary hematopoiesis.


Asunto(s)
Adenoviridae/genética , Factor VIII/genética , Expresión Génica , Vectores Genéticos , Animales , Secuencia de Bases , Cartilla de ADN , Humanos , Hígado/metabolismo , Macaca fascicularis , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción Genética
12.
Hum Gene Ther ; 6(2): 185-93, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7537539

RESUMEN

Hemophilia A is caused by blood coagulation factor VIII (FVIII) deficiency and is an attractive target for gene therapy. However, features of FVIII physiology, such as the instability of the mRNA and protein, have provided obstacles to the design of a feasible strategy for the transfer and expression of the human FVIII gene in vivo. We have constructed a recombinant adenoviral vector, Av1ALH81, that contains the human FVIII cDNA from which the B-domain has been deleted (BDD FVIII) and extensively characterized this vector in vitro and in vivo. In vitro, HepG2, human hepatoma cells, transduced with Av1ALH81 secreted high levels of biologically active human BDD FVIII measured by the Coatest bioassay (> 2,400 mU per 10(6) cells per 24 hr). Administration of Av1ALH81 to mice, via tail vein, resulted in expression of human BDD FVIII in the mouse plasma at levels averaging 307 +/- 93 ng/ml 1 week post-injection, measured by a sensitive human FVIII-specific ELISA. Normal FVIII levels in humans are 100-200 ng/ml, and therapeutic levels are as low as 10 ng/ml. Purification of the human FVIII from the mouse plasma, and subsequent Coatest analysis, revealed that the human FVIII produced in the mice was biologically active. In addition, the duration of FVIII expression in vivo was followed, and high-level FVIII expression was sustained over a period of several weeks. The finding that an adenoviral vector can mediate high-level expression of human FVIII in an animal model provides the basis for the development of gene therapy for hemophilia A.


Asunto(s)
Adenoviridae/genética , Factor VIII/genética , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Animales , ADN/análisis , Factor VIII/análisis , Factor VIII/biosíntesis , Factor VIII/aislamiento & purificación , Femenino , Humanos , Hígado/química , Ratones , Ratones Endogámicos C57BL , ARN/análisis , Eliminación de Secuencia/fisiología , Células Tumorales Cultivadas
13.
Hum Gene Ther ; 10(11): 1791-802, 1999 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-10446919

RESUMEN

Hemophilia A is the most common severe hereditary coagulation disorder and is caused by a deficiency in blood clotting factor VIII (FVIII). Canine hemophilia A represents an excellent large animal model that closely mimicks the human disease. In previous studies, treatment of hemophiliac dogs with an adenoviral vector encoding human FVIII resulted in complete correction of the coagulation defect and high-level FVIII expression [Connelly et al. (1996). Blood 88, 3846]. However, FVIII expression was short term, limited by a strong antibody response directed against the human protein. Human FVIII is highly immunogenic in dogs, whereas the canine protein is significantly less immunogenic. Therefore, sustained phenotypic correction of canine hemophilia A may require the expression of the canine protein. In this work, we have isolated the canine FVIII cDNA and generated an adenoviral vector encoding canine FVIII. We demonstrate expression of canine FVIII in hemophiliac mice at levels 10-fold higher than those of the human protein expressed from an analogous vector. Canine FVIII expression was sustained above human therapeutic levels (50 mU/ml) for at least 1 year in hemophiliac mice.


Asunto(s)
Adenoviridae/genética , Factor VIII/genética , Factor VIII/metabolismo , Vectores Genéticos , Hemofilia A/terapia , Animales , ADN Complementario/genética , Modelos Animales de Enfermedad , Perros , Estudios de Evaluación como Asunto , Técnicas de Transferencia de Gen , Terapia Genética , Humanos , Hígado/química , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción Genética
14.
Thromb Haemost ; 78(1): 31-6, 1997 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9198123

RESUMEN

Significant progress has been made on the development of gene therapy for the treatment of hemophilia A, a common bleeding disorder caused by subnormal levels of blood coagulation factor VIII (FVIII). Recent advances in gene transfer technology have enabled the expression of therapeutic to physiological levels of human FVIII in normal animals as well as hemophiliac mice and dogs. However, the in vivo persistence of FVIII expression was variable, ranging from one day to over five months. Despite recent advances in the development of hemophilia A gene transfer vectors, each still faces limitations to its clinical utility. Current research is focused on improving gene transfer vehicles and delivery methods to enable sustained clotting factor expression, treatment readministration, and circumvention of the host immune response to the treatment.


Asunto(s)
Factor VIII/genética , Terapia Genética , Hemofilia A/terapia , Células Cultivadas , Técnicas de Transferencia de Gen , Vectores Genéticos , Hemofilia A/genética , Humanos
15.
Thromb Haemost ; 81(2): 234-9, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10063998

RESUMEN

Adenoviral vectors provide a promising gene therapy system for the treatment of hemophilia A. Potent vectors encoding a human factor VIII (FVIII) cDNA were developed that mediated sustained FVIII expression in normal and hemophiliac mice and complete phenotypic correction of the bleeding disorder in hemophiliac mice and dogs (Connelly and Kaleko, Haemophilia 1998; 4: 380-8). However, these studies utilized vectors encoding a truncated version of the human FVIII cDNA lacking the B-domain (BDD FVIII). In this work, an adenoviral vector encoding the human full-length (FL) FVIII cDNA was generated and characterized. While functional FL FVIII was secreted in vitro, expression of the FL protein was not detected in the plasma of vector-treated hemophiliac mice. Unexpectedly, the FL FVIII vector-treated animals demonstrated phenotypic correction of the bleeding defect as measured by a tail-clip survival study. FL FVIII protein was visualized in the mouse livers using human FVIII-specific immunohistochemical analyses. These data demonstrate that adenoviral vector-mediated in vivo expression of BDD FVIII is more efficient than that of the FL protein and that phenotypic correction can occur in the absence of detectable levels of FVIII.


Asunto(s)
Adenovirus Humanos/genética , Factor VIII/genética , Terapia Genética , Vectores Genéticos/uso terapéutico , Hemofilia A/terapia , Animales , ADN Complementario/genética , Estudios de Evaluación como Asunto , Factor VIII/química , Vectores Genéticos/genética , Humanos , Hígado/química , Ratones , Ratones Noqueados , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/uso terapéutico , Células Tumorales Cultivadas
16.
Haemophilia ; 4(4): 380-8, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9873758

RESUMEN

Gene therapy for haemophilia A would represent a significant improvement over the current treatment by providing prophylactic expression of FVIII and correction of the coagulation defect. Furthermore, a gene therapy protocol allowing simple, infrequent vector administration may extend haemophilia treatment to remote locations world-wide that currently lack access to FVIII replacement therapy. Within the last half decade, significant progress has been made on the development of gene therapy for the treatment of haemophilia A. Recent achievements include high level clotting factor expression in mice, dogs, and monkeys as well as phenotypic correction in haemophiliac mice and dogs. With the efforts that are currently directed toward the improvement of gene transfer vectors and the development of technologies to enable sustained clotting factor expression, gene therapy for haemophilia A will ultimately become a reality.


Asunto(s)
Factor VIII/genética , Terapia Genética , Hemofilia A/genética , Hemofilia A/terapia , Animales , Perros , Factor VIII/uso terapéutico , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Ratones
17.
Cell ; 28(4): 801-11, 1982 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-6284374

RESUMEN

Postaggregative gene expression in Dictyostelium discoideum requires cell contact. Polyspecific monovalent antibodies (Fab) prepared from sera raised against membranes of aggregation- and postaggregation-stage cells were used to probe the cell interactions that induce rapid postaggregative synthesis of UDP-glucose pyrophosphorylase. When cells of strain V12M2 were dissociated after 8 hr of development and replated in the presence of immune Fab, both reaggregation and pyrophosphorylase synthesis were blocked. Fab neutralized by incubation with EDTA-high salt extracts of cells developed for 3 hr blocked pyrophosphorylase synthesis but not reaggregation. Therefore, some cell-surface components that regulate pyrophosphorylase synthesis (called E sites) are antigenically distinct from those required for reaggregation. The Fab provides a means to assay E sites during their purification. Addition of 10(-3) M cyclic AMP or cyclic GMP enabled the cells to bypass the blocking of E sites by Fab; pyrophosphorylase was synthesized in the absence of reaggregation. We hypothesize that E sites function by raising the level of intracellular cyclic AMP.


Asunto(s)
Membrana Celular/metabolismo , Dictyostelium/genética , Regulación de la Expresión Génica , Comunicación Celular , AMP Cíclico/farmacología , GMP Cíclico/farmacología , Dictyostelium/crecimiento & desarrollo , Ácido Edético/farmacología , Inducción Enzimática , Fragmentos Fab de Inmunoglobulinas/inmunología , UTP-Glucosa-1-Fosfato Uridililtransferasa/biosíntesis
18.
Haemophilia ; 5(3): 160-8, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10444282

RESUMEN

Hemophilia A is a severe bleeding disorder caused by a deficiency in blood coagulation factor VIII (FVIII). Adenoviral vectors containing a potent human FVIII expression cassette encoding a truncated FVIII cDNA were developed that mediated sustained FVIII expression in normal and haemophiliac mice and complete phenotypic correction of the bleeding disorder in haemophiliac mice and dogs (Connelly and Kaleko, Haemophilia, 1998; 4: 380-8). Here, we evaluated two E1/E2a/E3-deleted adenoviral vectors encoding human FVIII, one containing the full-length cDNA and the second containing a truncated cDNA lacking the B-domain. Viral vectors encoding the human full-length FVIII cDNA have not been described previously. Hepatocyte transduction was efficient and dose dependent, ranging from 50% to 100%. High levels of functional FVIII were secreted from transduced cells at amounts up to 6000 mU-1 10(6)cells-1 60 h. B-domain deleted FVIII was expressed at levels at least 8-fold higher than the full-length FVIII protein, whereas FVIII RNA levels were similar with both vectors. These data provide the first demonstration of FVIII adenoviral vector function in primary human cells and verify the potential clinical utility of adenoviral vectors for the treatment of haemophilia A.


Asunto(s)
Factor VIII/genética , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos , Hemofilia A/genética , Hemofilia A/terapia , Hígado/metabolismo , Adenoviridae , Animales , Secuencia de Bases , Células CHO , Cricetinae , ADN Complementario/genética , ADN Complementario/uso terapéutico , Perros , Factor VIII/biosíntesis , Expresión Génica , Hemofilia A/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular
19.
J Virol ; 72(8): 6875-9, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9658137

RESUMEN

Immunity to adenoviruses is an important hurdle to be overcome for successful gene therapy. The presence of antibodies to the capsid proteins prevents efficacious adenovirus vector administration in vivo. We tested whether immunity to a particular serotype of adenovirus (Ad5) may be overcome with a vector that encodes the hexon sequences from a different adenovirus serotype (Ad12). We successfully constructed an adenovirus vector with a chimeric Ad5-Ad12 hexon which was not neutralized by plasma from C57BL/6 mice immunized with Ad5. The vector was also capable of transducing the livers of C57BL/6 mice previously immunized with Ad5.


Asunto(s)
Adenovirus Humanos/inmunología , Proteínas de la Cápside , Cápside/inmunología , Vectores Genéticos/inmunología , Adenovirus Humanos/genética , Secuencia de Aminoácidos , Animales , Vectores Genéticos/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Transformación Genética
20.
J Cell Physiol ; 176(2): 392-401, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9648927

RESUMEN

Mouse P2A2-LISN myoblasts are transfected cells that overexpress the human type 1 insulin-like growth factor (IGF) receptor. Because the type 1 IGF receptor is the major binding site for both IGF-I and IGF-II, this cell line is an excellent model to determine the effect of multivalent cations on ligand binding specifically to this type of receptor. Competitive binding assays were performed to characterize IGF binding and Scatchard analysis to quantify affinity (Ka). 125I-IGF-I, 125I-IGF-II, and 125I-R3-IGF-I bind only to the type 1 IGF receptor on these cells. Zn2+ increased binding of the three ligands to the type 1 IGF receptor by 17 to 35%. Cd2+ significantly increased binding of 125I-IGF-I, although by only 8%. La3+ and Cr3+ did not effect binding. Au3+ decreased IGF binding by approximately 56%. Scatchard analysis produced nonlinear concave-down plots yielding binding constants for high and low affinity sites. Zn2+ increased the strength of only the high affinity sites. Au3+ decreased the affinity of both high and low affinity sites. Zn2+ increased binding with a half-maximal effect between 40 microM and 60 microM. Half-maximal dose of Au3+ was >130 microM. Zinc, gold, and cadmium bind to similar regions within proteins (a zinc-binding motif) and only these cations were found to affect receptor binding indicating similar mechanisms of action. Thus, multivalent cations may alter IGF binding to cell surface receptors ultimately controlling growth. Physiologically this may be especially important for the growth promoting effects of Zn2+.


Asunto(s)
Fibras Musculares Esqueléticas/química , Receptor IGF Tipo 1/metabolismo , Zinc/farmacología , Animales , Unión Competitiva/fisiología , Cadmio/farmacología , Cationes/farmacología , Células Cultivadas , Cromo/farmacología , Relación Dosis-Respuesta a Droga , Oro/farmacología , Factor I del Crecimiento Similar a la Insulina/análogos & derivados , Factor I del Crecimiento Similar a la Insulina/farmacología , Factor II del Crecimiento Similar a la Insulina/farmacología , Radioisótopos de Yodo , Lantano/farmacología , Ligandos , Ratones , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/citología , Ensayo de Unión Radioligante , Receptor IGF Tipo 1/agonistas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA