Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Haemophilia ; 22 Suppl 5: 31-5, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27405673

RESUMEN

INTRODUCTION: Haemophilia A is an X-linked bleeding disorder characterized by a deficiency of coagulation protein factor VIII (FVIII). A challenging complication of therapeutic FVIII infusions is the formation of neutralizing alloantibodies against the FVIII protein defined as inhibitors. The development of FVIII inhibitors drastically alters the quality of life of the patients and is associated with tremendous increases in morbidity as well as treatment costs. AIM: Current clinical immune tolerance induction protocols to reverse inhibitors are lengthy, costly and not effective in all patients. Prophylactic protocols to prevent inhibitor formation have not yet been developed in the clinical setting. However, there has been ample progress towards this goal in recent years in preclinical studies using animal models of haemophilia. METHODS: Here, we review the mechanisms that lead to inhibitor formation against FVIII and two promising new strategies for antigen-specific tolerance induction. RESULTS: CD4+ T cells play an important role in the FVIII-specific B cell response. Immune tolerance can be induced based on transplacental delivery of FVIII domains fused to Fc or on oral delivery of leaf cells from chloroplast transgenic crop plants. CONCLUSIONS: Recent literature suggests that prophylactic tolerance induction protocols for FVIII may be feasible in haemophilia A patients.


Asunto(s)
Hemofilia A/inmunología , Tolerancia Inmunológica , Animales , Anticuerpos Neutralizantes/sangre , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Modelos Animales de Enfermedad , Factor VIII/inmunología , Factor VIII/uso terapéutico , Femenino , Hemofilia A/tratamiento farmacológico , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo , Intercambio Materno-Fetal , Plantas Modificadas Genéticamente/inmunología , Embarazo
2.
Nat Genet ; 24(3): 257-61, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10700178

RESUMEN

Pre-clinical studies in mice and haemophilic dogs have shown that introduction of an adeno-associated viral (AAV) vector encoding blood coagulation factor IX (FIX) into skeletal muscle results in sustained expression of F.IX at levels sufficient to correct the haemophilic phenotype. On the basis of these data and additional pre-clinical studies demonstrating an absence of vector-related toxicity, we initiated a clinical study of intramuscular injection of an AAV vector expressing human F.IX in adults with severe haemophilia B. The study has a dose-escalation design, and all patients have now been enrolled in the initial dose cohort (2 x 10(11) vg/kg). Assessment in the first three patients of safety and gene transfer and expression show no evidence of germline transmission of vector sequences or formation of inhibitory antibodies against F.IX. We found that the vector sequences are present in muscle by PCR and Southern-blot analyses of muscle biopsies and we demonstrated expression of F.IX by immunohistochemistry. We observed modest changes in clinical endpoints including circulating levels of F.IX and frequency of FIX protein infusion. The evidence of gene expression at low doses of vector suggests that dose calculations based on animal data may have overestimated the amount of vector required to achieve therapeutic levels in humans, and that the approach offers the possibility of converting severe haemophilia B to a milder form of the disease.


Asunto(s)
Dependovirus/genética , Factor IX/genética , Terapia Genética , Vectores Genéticos/uso terapéutico , Hemofilia B/terapia , Músculo Esquelético/metabolismo , Adulto , Anciano , Pruebas de Coagulación Sanguínea , Southern Blotting , Factor IX/análisis , Expresión Génica , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Hemofilia B/genética , Humanos , Inyecciones Intramusculares , Masculino , Músculo Esquelético/virología , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Resultado del Tratamiento
3.
Nat Med ; 5(1): 56-63, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9883840

RESUMEN

Hemophilia B is a severe X-linked bleeding diathesis caused by the absence of functional blood coagulation factor IX, and is an excellent candidate for treatment of a genetic disease by gene therapy. Using an adeno-associated viral vector, we demonstrate sustained expression (>17 months) of factor IX in a large-animal model at levels that would have a therapeutic effect in humans (up to 70 ng/ml, adequate to achieve phenotypic correction, in an animal injected with 8.5x10(12) vector particles/kg). The five hemophilia B dogs treated showed stable, vector dose-dependent partial correction of the whole blood clotting time and, at higher doses, of the activated partial thromboplastin time. In contrast to other viral gene delivery systems, this minimally invasive procedure, consisting of a series of percutaneous intramuscular injections at a single timepoint, was not associated with local or systemic toxicity. Efficient gene transfer to muscle was shown by immunofluorescence staining and DNA analysis of biopsied tissue. Immune responses against factor IX were either absent or transient. These data provide strong support for the feasibility of the approach for therapy of human subjects.


Asunto(s)
Dependovirus , Factor IX/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Hemofilia B/terapia , Animales , ADN Viral/análisis , Dependovirus/genética , Modelos Animales de Enfermedad , Perros , Factor IX/inmunología , Expresión Génica , Hemofilia B/inmunología , Humanos , Inyecciones Intramusculares , Masculino , Factores de Tiempo , Células Tumorales Cultivadas
4.
Gene Ther ; 17(3): 295-304, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19907498

RESUMEN

Viral vectors are potent gene delivery platforms used for the treatment of genetic and acquired diseases. However, just as viruses have evolved to infect cells efficiently, the immune system has evolved to fight off what it perceives as invading pathogens. Therefore, innate immunity and antigen-specific adaptive immune responses against vector-derived antigens reduce the efficacy and stability of in vivo gene transfer. In addition, a number of vectors are derived from parent viruses that humans encounter through natural infection, resulting in preexisting antibodies and possibly in memory responses against vector antigens. Similarly, antibody and T-cell responses may be directed against therapeutic gene products that often differ from the endogenous nonfunctional or absent protein that is being replaced. As details and mechanisms of such immune reactions are uncovered, novel strategies are being developed, and vectors are being specifically engineered to avoid, suppress or manipulate the response, ideally resulting in sustained expression and immune tolerance to the transgene product. This review provides a summary of our current knowledge of the interactions between the immune system adeno-associated virus, adenoviral and lentiviral vectors, and their transgene products.


Asunto(s)
Adenoviridae/inmunología , Dependovirus/inmunología , Terapia Genética , Vectores Genéticos/inmunología , Lentivirus/inmunología , Animales , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Proteínas del Sistema Complemento/inmunología , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Ratones , Linfocitos T/inmunología , Receptor Toll-Like 9/inmunología
5.
Biotechniques ; 34(1): 184-9, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12545558

RESUMEN

Adeno-associated viral (AAV) vectors are used for in vivo gene transfer in a number of preclinical models of genetic diseases (including large-animal models) and are currently being tested in clinical trials for treatment of hemophilia B and cystic fibrosis. Protocols for production of AAV vectors in a helper virus-free system are available and are based on transient transfection of HEK-293 cells with multiple plasmids. Scale-up of vector production has been labor intensive and inefficient because of a lack of larger culture vessels suitable for growth of adherent cells, large-scale transfection, and vector production. Here we report efficient production of AAV vector in roller bottles, which represents a 10-fold scale-up from the conventional flask or plate method. Optimized production yielded greater than 10(13) vector genomes per bottle and was as cost effective as published protocols using plates. Successful vector production by this method was dependent on optimization of transfection by calcium phosphate precipitation, of monitoring of cell growth (by measurement of glucose consumption), of cell culture conditions, and CO2/air exchange with the culture vessel.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Dependovirus/genética , Vectores Genéticos/biosíntesis , Vectores Genéticos/aislamiento & purificación , Transfección/instrumentación , Recuento de Células , Técnicas de Cultivo de Célula/métodos , Línea Celular , Dependovirus/crecimiento & desarrollo , Estudios de Factibilidad , Vectores Genéticos/análisis , Vectores Genéticos/síntesis química , Glucosa/metabolismo , Humanos , Riñón/embriología , Riñón/fisiología , Control de Calidad , Recombinación Genética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Transfección/métodos
6.
Am J Pharmacogenomics ; 1(2): 137-44, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-12174674

RESUMEN

The year 2000 saw the first successful treatment of a genetic disorder by gene therapy. Pediatric patients with X-linked severe combined immunodeficiency disorder (SCID-X1) received autologous CD34+ hematopoietic cells following ex vivo gene transfer using a retroviral vector, with subsequent demonstration of improved immune responses. A number of preclinical and clinical studies have been conducted with the aim of developing gene therapy for hemophilia, Fanconi anemia, sickle cell disease, beta-thalassemia, chronic granulomatous disease, and other inherited hematological disorders. The greatest advances in novel approaches toward treatment of hematological disorders have been made in hemophilia, with 3 current phase I clinical trials ongoing. Two trials are investigating the safety and feasibility of utilizing either an ex vivo, non-viral gene transfer technique or an intravenous infusion of a retroviral vector to treat adults with severe hemophilia A (factor VIII deficiency). The third study involves intramuscular administration of an adeno-associated viral (AAV) vector for expression of factor IX in adult patients with hemophilia B. Results from this study and from preclinical studies preceding the trial demonstrate that it is possible to safely administer high doses of a viral vector in vivo.


Asunto(s)
Terapia Genética/métodos , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/terapia , Animales , Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Terapia Genética/estadística & datos numéricos , Terapia Genética/tendencias , Enfermedad Granulomatosa Crónica/genética , Enfermedad Granulomatosa Crónica/terapia , Hemoglobinopatías/genética , Hemoglobinopatías/terapia , Hemofilia A/genética , Hemofilia A/terapia , Humanos , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/terapia
7.
J Thromb Haemost ; 9(8): 1524-33, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21585650

RESUMEN

BACKGROUND: Formation of inhibitory antibodies is a frequent and serious complication of factor (F) VIII replacement therapy for the X-linked bleeding disorder hemophilia A. Similarly, hemophilia A mice develop high-titer inhibitors to recombinant human FVIII after a few intravenous injections. OBJECTIVE: Using the murine model, the study sought to develop a short regimen capable of inducing tolerance to FVIII. METHODS: A 1-month immunomodulatory protocol, consisting of FVIII administration combined with oral delivery of rapamycin, was developed. RESULTS: The protocol effectively prevented formation of inhibitors to FVIII upon subsequent intravenous treatment (weekly for 3.5 months). Control mice formed high-titer inhibitors and had CD4(+) T effector cell responses characterized by expression of IL-2, IL-4 and IL-6. Tolerized mice instead had a CD4(+)CD25(+)FoxP3(+) T cell response to FVIII that suppressed antibody formation upon adoptive transfer, indicating a shift from Th2 to Treg if FVIII antigen was introduced to T cells during inhibition with rapamycin. CD4(+) T cells from tolerized mice also expressed TGF-ß1 and CTLA4, but not IL-10. The presence of FVIII antigen during the time of rapamycin administration was required for specific tolerance induction. CONCLUSIONS: The study shows that a prophylactic immune tolerance protocol for FVIII can be developed using rapamycin, a drug that is already widely in clinical application. Immune suppression with rapamycin was mild and highly transient, as the mice regained immune competence within a few weeks.


Asunto(s)
Anticuerpos/sangre , Linfocitos T CD4-Positivos/efectos de los fármacos , Coagulantes/administración & dosificación , Factor VIII/administración & dosificación , Hemofilia A/tratamiento farmacológico , Tolerancia Inmunológica/efectos de los fármacos , Inmunosupresores/administración & dosificación , Sirolimus/administración & dosificación , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Antígeno CTLA-4/metabolismo , Células Cultivadas , Coagulantes/inmunología , Modelos Animales de Enfermedad , Esquema de Medicación , Factor VIII/inmunología , Factores de Transcripción Forkhead/metabolismo , Hemofilia A/sangre , Hemofilia A/inmunología , Humanos , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Recombinantes/administración & dosificación , Factores de Tiempo , Factor de Crecimiento Transformador beta1/metabolismo
10.
J Thromb Haemost ; 7 Suppl 1: 88-91, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19630776

RESUMEN

The immune response to coagulation factors VIII or IX, in particular formation of inhibitory antibodies, complicates treatment of hemophilia. Therefore, a number of recent studies in animal models have explored novel approaches toward induction of immune tolerance in protein or gene replacement therapy. Strong evidence has emerged that regulatory T cells (Treg) are an important component of the mechanism by which tolerance is maintained and inhibitor formation, a T help dependent response, is prevented. Limited data in patients also support this concept. In particular, CD4+ CD25+ FoxP3+ Treg, whether naturally occurring or induced, have been invoked in suppression of antibody and of cytotoxic T lymphocyte responses to the therapeutic clotting factor. This review summarizes the data on this emerging concept of Treg-mediated regulation of the immune response in treatment of hemophilia, strategies and mechanisms of Treg induction and function, and the implications for development of immune tolerance protocols.


Asunto(s)
Factores de Coagulación Sanguínea/inmunología , Tolerancia Inmunológica/fisiología , Linfocitos T Reguladores/fisiología , Hemofilia A/inmunología , Hemofilia A/terapia , Humanos
11.
J Thromb Haemost ; 7(9): 1523-32, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19583824

RESUMEN

BACKGROUND: Gene and protein replacement therapies for inherited protein deficiencies such as hemophilia or lysosomal storage disorders are limited by deleterious immune responses directed against their respective therapeutic proteins. Therefore, the development of protocols preventing such responses is key to providing successful long-term therapy. OBJECTIVES: We sought to develop a protocol, utilizing a drug/peptide cocktail, that would effectively shift the antigen-specific CD4+ T-cell population, tipping the balance from effector T cells (Teffs) towards regulatory T cells (Tregs). METHODS: Treg-deficient (DO11.10-tg Rag2(-/-)) BALB/c mice were used to screen for an optimal protocol addressing the aforementioned goal and to study the mechanisms underlying in vivo changes in T-cell populations. Muscle-directed gene transfer to hemophilia B mice was also performed in order to test the optimal protocol in a therapeutically relevant setting. RESULTS: Specific antigen administration (4-week repeated dosing) combined with rapamycin and interleukin-10 led to substantial reductions in Teffs, via activation-induced cell death, and induced CD4+CD25+FoxP3+ Tregs to a large extent in multiple organs. The proportion of apoptotic T cells also increased over time, whereas Teffs and Tregs were differentially affected. When applied to a model of protein deficiency (gene therapy for hemophilia B), the protocol successfully prevented inhibitor formation, whereas non-specific immunosuppression was only marginally effective. CONCLUSIONS: It is feasible to provide a short-term, prophylactic protocol allowing for the induction of immune tolerance. This protocol may provide a marked advance in efforts seeking to improve clinical outcomes in disorders involving therapeutic protein replacement.


Asunto(s)
Antígenos/química , Factor IX/genética , Terapia Genética/métodos , Hemofilia A/genética , Tolerancia Inmunológica , Sirolimus/farmacología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Factores de Transcripción Forkhead/biosíntesis , Hemofilia A/metabolismo , Humanos , Interleucina-10/metabolismo , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H
12.
J Thromb Haemost ; 6(2): 339-45, 2008 02.
Artículo en Inglés | MEDLINE | ID: mdl-18036190

RESUMEN

BACKGROUND: Activated factor X (FXa) is a vitamin K-dependent serine protease that plays a pivotal role in blood coagulation by converting prothrombin to thrombin. There are no reports of humans with complete deficiency of FX, and knockout of murine F10 is embryonic or perinatal lethal. OBJECTIVE: We sought to generate a viable mouse model of FX deficiency. METHODS: We used a socket-targeting construct to generate F10-knockout mice by eliminating F10 exon 8 (knockout allele termed F10(tm1Ccmt), abbreviated as '-'; wild-type '+'), and a plug-targeting construct to generate mice expressing a FX variant with normal antigen levels but low levels of FX activity [4-9% normal in humans carrying the defect, Pro343-->Ser, termed FX Friuli (mutant allele termed F10(tm2Ccmt), abbreviated as F)]. RESULTS: F10 knockout mice exhibited embryonic or perinatal lethality. In contrast, homozygous Friuli mice [F10 (F/F)] had FX activity levels of approximately 5.5% (sufficient to rescue both embryonic and perinatal lethality), but developed age-dependent iron deposition and cardiac fibrosis. Interestingly, F10 (-/F) mice with FX activity levels of 1-3% also showed complete rescue of lethality. Further study of this model provides evidence supporting a role of maternal FX transfer in the embryonic survival. CONCLUSIONS: We demonstrate that, while complete absence of FX is incompatible with murine survival, minimal FX activity as low as 1-3% is sufficient to rescue the lethal phenotype. This viable low-FX mouse model will facilitate the development of FX-directed therapies as well as investigation of the FX role in embryonic development.


Asunto(s)
Deficiencia del Factor X/genética , Factor X/genética , Impresión Genómica/genética , Ratones Transgénicos/genética , Modelos Animales , Sustitución de Aminoácidos , Animales , Cardiomiopatías/etiología , Exones/genética , Deficiencia del Factor X/complicaciones , Femenino , Muerte Fetal/genética , Fibrosis , Marcación de Gen/métodos , Genes Letales , Prueba de Complementación Genética , Genotipo , Hemosiderosis/etiología , Humanos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos/sangre , Miocardio/patología
13.
Gene Ther ; 12(19): 1453-64, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15889137

RESUMEN

The risk of an immune response to the coagulation factor IX (F.IX) transgene product is a concern in gene therapy for the X-linked bleeding disorder hemophilia B. In order to investigate the mechanism of F.IX-specific lymphocyte activation in the context of adeno-associated viral (AAV) gene transfer to skeletal muscle, we injected AAV-2 vector expressing human F.IX (hF.IX) into outbred immune-competent mice. Systemic hF.IX levels were transiently detected in the circulation, but diminished concomitant with activation of CD4+ T and B cells. ELISPOT assays documented robust responses to hF.IX in the draining lymph nodes of injected muscle by day 14. Formation of inhibitory antibodies to hF.IX was observed over a wide range of vector doses, with increased doses causing stronger immune responses. A prolonged inflammatory reaction in muscle started at 1.5-2 months, but ultimately failed to eliminate transgene expression. By 1.5 months, hF.IX antigen re-emerged in circulation in approximately 70% of animals injected with high vector dose. Hepatic gene transfer elicited only infrequent and weaker immune responses, with higher vector doses causing a reduction in T-cell responses to hF.IX. In summary, the data document substantial influence of target tissue, local antigen presentation, and antigen levels on lymphocyte responses to F.IX.


Asunto(s)
Anticuerpos Bloqueadores/inmunología , Dependovirus/genética , Factor IX/inmunología , Terapia Genética/efectos adversos , Hemofilia B/terapia , Músculo Esquelético/inmunología , Adenovirus Humanos/genética , Animales , Formación de Anticuerpos , Linfocitos B/inmunología , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Hemofilia B/inmunología , Humanos , Inyecciones Intramusculares , Interferón gamma/inmunología , Interleucina-4/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos , Linfocitos T Colaboradores-Inductores/inmunología , Transducción Genética/métodos , Transgenes
14.
Curr Opin Hematol ; 5(5): 321-6, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9776210

RESUMEN

The aim of gene therapy for hemophilia is the stable introduction and expression of a gene encoding functional blood coagulation factor VIII or IX. Although there are as yet no published studies demonstrating long-term expression of therapeutic levels in large animal models of the disease, there have been several reports over the past year of sustained expression of therapeutic levels of clotting factors in small animals, and some of these strategies are currently being applied to hemophilic dogs. Recent advances include optimized retroviral gene transfer, improved adenoviral vectors for high levels of sustained expression of factor VIII in mice, stable therapeutic levels of factor IX expression in mice after transduction of muscle or liver with adenoassociated virus vector, as well as new nonviral gene delivery strategies. Finally, several important mouse and dog models of hemophilia have been characterized during the past year.


Asunto(s)
Terapia Genética , Hemofilia A/terapia , Animales , Modelos Animales de Enfermedad , Factor IX/genética , Factor IX/uso terapéutico , Factor VIII/genética , Factor VIII/uso terapéutico , Técnicas de Transferencia de Gen , Hemofilia A/genética , Humanos
15.
Appl Environ Microbiol ; 64(6): 2229-31, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9603839

RESUMEN

A differentially expressed gpdA cDNA clone was isolated from NaCl-adapted Aspergillus nidulans (FGSC359) and identified as glyceraldehyde-3-phosphate dehydrogenase (gpdA) on the basis of its nucleotide sequence. The level of gpdA RNA substantially increased in cultures gradually adapted to NaCl but was greatly reduced in cultures exposed briefly to a high concentration of NaCl. A pyrG auxotroph of A. nidulans (A773) was cotransformed with a gpdA-uidA construct and a plasmid containing the Neurospora crassa pyr4 gene as a selectable marker. One pyrG+ beta-glucuronidase-positive (GUS+) transformant was selected, and stable integration of the gpdA-uidA construct into the genome was confirmed by Southern blot analysis. Gradual adaptation to increasing concentrations of NaCl resulted in an increase in GUS activity to 2.7-fold. GUS activity was reduced after a 2-h exposure of an unadapted culture to 2 M NaCl but gradually increased to a maximum of twofold after 24 h. GUS activity also increased by 8.4-fold in Na2SO4-adapted cultures, 4.9-fold in polyethylene glycol-adapted cultures, and 7.5-fold in KCl-adapted cultures. These results are consistent with the hypothesis that the A. nidulans gpdA promoter is transcriptionally activated by osmotic signals.


Asunto(s)
Aspergillus nidulans/enzimología , Aspergillus nidulans/genética , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Regiones Promotoras Genéticas , Adaptación Fisiológica , Fusión Artificial Génica , Aspergillus nidulans/metabolismo , Genes Fúngicos , Genes Reporteros , Glucuronidasa/genética , Ósmosis , Transducción de Señal , Cloruro de Sodio , Activación Transcripcional , Transformación Genética
16.
Curr Genet ; 27(5): 460-5, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7586033

RESUMEN

Replicative plasmids pP01 and pP02, recovered from Pleurotus ostreatus transformants, contain an insert of bacteriophage origin. These plasmids have been amplified by the polymerase chain reaction (PCR) and have been shown to represent a low-grade component in the initial preparation of the vector pAN7-1. The pP01 and pP02 plasmids share an insert (P01A) of virtual identity with a SmaI-BamHI genomic fragment of P 1 bacteriophage and retain remnants of a polylinker at the 5' end of this fragment. Such an insert undoubtedly represents an in vitro-generated event and did not arise, as suggested previously, by recombination of pAN7-1 with the P. ostreatus genome. The P. ostreatus transformants, however, do select for the minority pP0 plasmid, apparently recognizing the P01A insert as a heterologous or surrogate replicon.


Asunto(s)
Bacteriófagos/genética , Plásmidos/genética , Polyporaceae/genética , Aspergillus/genética , Secuencia de Bases , Farmacorresistencia Microbiana , Higromicina B/farmacología , Datos de Secuencia Molecular , Selección Genética , Transformación Genética
17.
Appl Microbiol Biotechnol ; 47(4): 368-72, 1997 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9163950

RESUMEN

A gene for a synthetic protein-based polymer, G-(VPGVG)119-VPGV, coding for the EG-120mer (elastomer), was cloned into a fungal expression vector to allow constitutive expression of the polymer controlled by the gpdA (glyceraldehyde-3-phosphate dehydrogenase) promoter sequence of Aspergillus nidulans. Stable transformants of A. nidulans showed plasmid integration with varying copy number when analyzed by Southern-blot hybridization. Expression of the synthetic gene was demonstrated by Northern-blot hybridization. However, the translational efficiency for production of the polymer polypeptide was low, presumably because of certain codons in the polymer gene (CCG and GUA) that are rarely used by A. nidulans. Partial purification by reversible phase transition followed by sodium dodecyl sulfate/polyacrylamide gel electrophoresis revealed the presence of polymer protein in a transformant that contained multiple copies of the polymer gene. This study represents the first attempt to express a synthetic gene (with no natural analog) in a fungus.


Asunto(s)
Aspergillus nidulans/genética , Aspergillus nidulans/metabolismo , Regulación Fúngica de la Expresión Génica , Goma/metabolismo , Northern Blotting , Southern Blotting , Clonación Molecular , ADN de Hongos/análisis , Electroforesis en Gel de Poliacrilamida , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Plásmidos , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , ARN de Hongos/análisis , Transformación Genética
18.
Mol Ther ; 4(3): 192-200, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11545609

RESUMEN

The X-linked bleeding disorder hemophilia B is caused by absence of functional blood coagulation factor IX (F9) and can be treated by adeno-associated viral (AAV) mediated gene transfer to skeletal muscle. The safety of this approach is currently being evaluated in a phase I clinical trial. Efficacy of this and several other gene therapy strategies has been addressed in hemophilia B dogs, an important preclinical model of the disease. While previously published data demonstrated sustained expression of canine F9 in dogs with a missense mutation in the gene F9, we show here that AAV-mediated canine F9 gene transfer to skeletal muscle of hemophilia B dogs carrying a null mutation of F9 (causing an early stop codon and an unstable mRNA) results in induction of inhibitory anti-canine F9 at comparable vector doses (1 x 10(12) vector genomes/kg). Thus, the risk of inhibitor formation following AAV-mediated F9 gene therapy may be influenced by the nature of the underlying mutation in F9. Transient immune suppression with cyclophosphamide at the time of vector administration blocked formation of anti-canine F9 antibodies in the one animal treated with this approach. Treatment with this combination of gene transfer and transient immune modulation has resulted in sustained expression (>8 months) of canine F9 at levels sufficient for partial correction of coagulation parameters.


Asunto(s)
Factor IX/uso terapéutico , Eliminación de Gen , Terapia Genética/métodos , Hemofilia B/genética , Hemofilia B/terapia , Inmunosupresores/farmacología , Músculo Esquelético/metabolismo , Adenoviridae/genética , Animales , Anticuerpos/inmunología , Western Blotting , Ciclofosfamida/farmacología , Enfermedades de los Perros/genética , Enfermedades de los Perros/terapia , Perros , Factor IX/genética , Factor IX/inmunología , Factor IX/farmacología , Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Hemofilia B/inmunología , Hemofilia B/veterinaria , Inyecciones Intramusculares , Masculino , Factores de Tiempo
19.
Blood ; 95(8): 2536-42, 2000 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-10753832

RESUMEN

Hemophilia B is caused by the absence of functional coagulation factor IX (F.IX) and represents an important model for treatment of genetic diseases by gene therapy. Recent studies have shown that intramuscular injection of an adeno-associated viral (AAV) vector into mice and hemophilia B dogs results in vector dose-dependent, long-term expression of biologically active F.IX at therapeutic levels. In this study, we demonstrate that levels of expression of approximately 300 ng/mL (6% of normal human F.IX levels) can be reached by intramuscular injection of mice using a 2- to 4-fold lower vector dose (1 x 10(11) vector genomes/mouse, injected into 4 intramuscular sites) than previously described. This was accomplished through the use of an improved expression cassette that uses the cytomegalovirus (CMV) immediate early enhancer/promoter in combination with a 1.2-kilobase portion of human skeletal actin promoter. These results correlated with enhanced levels of F.IX transcript and secreted F.IX protein in transduced murine C2C12 myotubes. Systemic F.IX expression from constructs containing the CMV enhancer/promoter alone was 120 to 200 ng/mL in mice injected with 1 x 10(11) vector genomes. Muscle-specific promoters performed poorly for F.IX transgene expression in vitro and in vivo. However, the incorporation of a sequence from the alpha-skeletal actin promoter containing at least 1 muscle-specific enhancer and 1 enhancer-like element further improved muscle-derived expression of F.IX from a CMV enhancer/promoter-driven expression cassette over previously published results. These findings will allow the design of a clinical protocol for therapeutic levels of F.IX expression with lower vector doses, thus enhancing efficacy and safety of the protocol. (Blood. 2000;95:2536-2542)


Asunto(s)
Factor IX/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Actinas/genética , Animales , Citomegalovirus/genética , Perros , Elementos de Facilitación Genéticos/genética , Factor IX/biosíntesis , Expresión Génica , Humanos , Ratones , Músculo Esquelético
20.
Mol Ther ; 4(3): 201-10, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11545610

RESUMEN

The safety of several gene therapy approaches for treatment of the severe, X-linked bleeding disorder hemophilia is currently being evaluated in early phase clinical trials. One strategy seeks to correct deficiency of functional coagulation factor IX (hemophilia B) by intramuscular (IM) administration of an adeno-associated viral (AAV) vector. A potentially serious complication of any treatment for hemophilia is formation of inhibitory antibodies against the coagulation factor protein, a risk that increases in the setting of null mutations in the factor IX gene (F9). Here, we describe hemophilia B mice with a large F9 deletion that form inhibitors within 1 to 2 months after IM administration of an AAV vector expressing mouse F9 or after repeated intravenous infusion of mouse F9 concentrate. In both cases, inhibitors are primarily IgG1 immunoglobulins representing a Th2-driven humoral immune response. We further demonstrate that anti-mouse F9 antibody formation in the gene-based approach can be reduced by transient immune modulation at the time of vector administration. Moreover, this maneuver resulted in complete absence of anti-mouse F9 and sustained expression of functional mouse F9 in some hemophilia B mice, particularly in those animals treated with the immunosuppressive drug cyclophosphamide. These data have direct relevance for design of clinical trials and strategies aimed at avoiding immune responses against a secreted transgene product.


Asunto(s)
Dependovirus/genética , Factor IX/genética , Factor IX/inmunología , Eliminación de Gen , Terapia Genética/métodos , Hemofilia B/genética , Hemofilia B/inmunología , Animales , Anticuerpos/inmunología , Células CHO , Cricetinae , Ciclofosfamida/farmacología , Factor IX/administración & dosificación , Factor IX/uso terapéutico , Expresión Génica/efectos de los fármacos , Vectores Genéticos/genética , Hemofilia B/tratamiento farmacológico , Hemofilia B/terapia , Inmunosupresores/farmacología , Inyecciones Intramusculares , Inyecciones Intravenosas , Ratones , Ratones Endogámicos C57BL , Tiempo de Tromboplastina Parcial , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Riesgo , Factores de Tiempo , Transgenes/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda