Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Ethn Dis ; 27(3): 249-256, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28811736

RESUMO

OBJECTIVE: To report baseline characteristics of junior-level faculty participants in the Summer Institute Programs to Increase Diversity (SIPID) and the Programs to Increase Diversity among individuals engaged in Health-Related Research (PRIDE), which aim to facilitate participants' career development as independent investigators in heart, lung, blood, and sleep research. DESIGN AND SETTING: Junior faculty from groups underrepresented in the biomedical-research workforce attended two, 2-3 week, annual summer research-education programs at one of six sites. Programs provided didactic and/or laboratory courses, workshops to develop research, writing and career-development skills, as well as a mentoring component, with regular contact maintained via phone, email and webinar conferences. Between summer institutes, trainees participated in a short mid-year meeting and an annual scientific meeting. Participants were surveyed during and after SIPID/PRIDE to evaluate program components. PARTICIPANTS: Junior faculty from underrepresented populations across the United States and Puerto Rico participated in one of three SIPID (2007-2010) or six PRIDE programs (2011-2014). RESULTS: Of 204 SIPID/PRIDE participants, 68% were female; 67% African American and 27% Hispanic/Latino; at enrollment, 75% were assistant professors and 15% instructors, with most (96%) on non-tenure track. Fifty-eight percent had research doctorates (PhD, ScD) and 42% had medical (MD, DO) degrees. Mentees' feedback about the program indicated skills development (eg, manuscript and grant writing), access to networking, and mentoring were the most beneficial elements of SIPID and PRIDE programs. Grant awards shifted from primarily mentored research mechanisms to primarily independent investigator awards after training. CONCLUSIONS: Mentees reported their career development benefited from SIPID and PRIDE participation.


Assuntos
Pesquisa Biomédica/organização & administração , Docentes de Medicina , Tutoria/métodos , Mentores , National Heart, Lung, and Blood Institute (U.S.) , Desenvolvimento de Programas , Feminino , Humanos , Masculino , Estados Unidos
2.
Blood ; 119(23): 5532-42, 2012 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-22368271

RESUMO

To better understand cellular basis of hemophilia, cell types capable of producing FVIII need to be identified. We determined whether bone marrow (BM)-derived cells would produce cells capable of synthesizing and releasing FVIII by transplanting healthy mouse BM into hemophilia A mice. To track donor-derived cells, we used genetic reporters. Use of multiple coagulation assays demonstrated whether FVIII produced by discrete cell populations would correct hemophilia A. We found that animals receiving healthy BM cells survived bleeding challenge with correction of hemophilia, although donor BM-derived hepatocytes or endothelial cells were extremely rare, and these cells did not account for therapeutic benefits. By contrast, donor BM-derived mononuclear and mesenchymal stromal cells were more abundant and expressed FVIII mRNA as well as FVIII protein. Moreover, injection of healthy mouse Kupffer cells (liver macrophage/mononuclear cells), which predominantly originate from BM, or of healthy BM-derived mesenchymal stromal cells, protected hemophilia A mice from bleeding challenge with appearance of FVIII in blood. Therefore, BM transplantation corrected hemophilia A through donor-derived mononuclear cells and mesenchymal stromal cells. These insights into FVIII synthesis and production in alternative cell types will advance studies of pathophysiological mechanisms and therapeutic development in hemophilia A.


Assuntos
Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Fator VIII/metabolismo , Hemofilia A/cirurgia , Animais , Células da Medula Óssea/citologia , Células Endoteliais/citologia , Hemofilia A/metabolismo , Humanos , Células de Kupffer/transplante , Transplante de Células-Tronco Mesenquimais , Camundongos , Camundongos Endogâmicos C57BL
4.
Mol Ther ; 19(3): 442-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21081906

RESUMO

Developing adeno-associated viral (AAV)-mediated gene therapy for hemophilia A (HA) has been challenging due to the large size of the factor VIII (FVIII) complementary DNA and the concern for the development of inhibitory antibodies to FVIII in HA patients. Here, we perform a systematic study in HA dogs by delivering a canine FVIII (cFVIII) transgene either as a single chain or two chains in an AAV vector. An optimized cFVIII single chain delivered using AAV serotype 8 (AAV8) by peripheral vein injection resulted in a dose-response with sustained expression of FVIII up to 7% (n = 4). Five HA dogs administered two-chain delivery using either AAV8 or AAV9 via the portal vein expressed long-term, vector dose-dependent levels of FVIII activity (up to 10%). In the two-chain approach, circulating cFVIII antigen levels were more than fivefold higher than activity. Notably, no long-term immune response to FVIII was observed in any of the dogs (1/9 dogs had a transient inhibitor). Long-term follow-up of the dogs showed a remarkable reduction (>90%) of bleeding episodes in a combined total of 24 years of observation. These data demonstrate that both approaches are safe and achieve dose-dependent therapeutic levels of FVIII expression, which supports translational studies of AAV-mediated delivery for HA.


Assuntos
Dependovirus , Fator VIII , Terapia Genética , Vetores Genéticos , Hemofilia A , Fígado , Animais , Dependovirus/genética , Modelos Animais de Doenças , Cães , Fator VIII/genética , Fator VIII/metabolismo , Regulação da Expressão Gênica , Ordem dos Genes , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Hemofilia A/imunologia , Hemofilia A/prevenção & controle , Hemofilia A/terapia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Resultado do Tratamento
5.
Mol Ther ; 18(11): 1907-16, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20700109

RESUMO

Delivery of genes that are larger than the wild-type adeno-associated virus (AAV) 4,681 nucleotide genome is inefficient using AAV vectors. We previously demonstrated in vitro that concurrent proteasome inhibitor (PI) treatment improves transduction by AAV vectors encoding oversized transgenes. In this study, an AAV vector with a 5.6 kilobase (kb) factor VIII expression cassette was used to test the effect of an US Food and Drug Administration-approved PI (bortezomib) treatment concurrent with vector delivery in vivo. Intrahepatic vector delivery resulted in factor VIII expression that persisted for >1 year in hemophilia mice. Single-dose bortezomib given with AAV2 or AAV8 factor VIII vector enhanced expression on average ~600 and ~300%, respectively. Moreover, coadministration of AAV8.canineFVIII (1 × 10(13) vg/kg) and bortezomib in hemophilia A dogs (n = 4) resulted in normalization of the whole blood clotting time (WBCT) and 90% reduction in hemorrhages for >32 months compared to untreated hemophilia A dogs (n = 3) or dogs administered vector alone (n = 3). Demonstration of long-term phenotypic correction of hemophilia A dogs with combination adjuvant bortezomib and AAV vector expressing the oversized transgene establishes preclinical studies that support testing in humans and provides a working paradigm to facilitate a significant expansion of therapeutic targets for human gene therapy.


Assuntos
Ácidos Borônicos/farmacologia , Dependovirus/genética , Modelos Animais de Doenças , Terapia Genética , Genoma Viral , Hemofilia B/genética , Inibidores de Proteases/farmacologia , Pirazinas/farmacologia , Animais , Coagulação Sanguínea/efeitos dos fármacos , Bortezomib , Núcleo Celular/metabolismo , Terapia Combinada , Cães , Fator IX/fisiologia , Fator VIII/fisiologia , Feminino , Vetores Genéticos , Hemofilia B/terapia , Humanos , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transgenes/fisiologia
6.
Mol Ther ; 17(3): 417-24, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19127250

RESUMO

Hemophilia A gene therapy using recombinant adenovirus-associated virus (AAV) vectors has been hampered by the size of the factor VIII (FVIII) cDNA. Previously, splitting the FVIII coding sequence into a heavy-chain (HC) fragment and a light-chain (LC) fragment for dual recombinant AAV vector delivery has been successfully explored. However, the main disadvantage of this approach is a "chain imbalance" problem in which LC secretion is approximately 1-2 logs higher than that of HC, and therefore, the majority of protein synthesized is nonfunctional. To improve HC secretion, we constructed alternate FVIII HCs based on our observation that LC facilitates HC secretion. To our surprise, most of the new HC molecules exhibited enhanced expression over the traditional HC molecule (HC(745)). The optimized HC mutein, HC(HL), including additional acidic-region-3 (ar3) sequences, exhibited three- to fivefold higher activity in both enzyme-linked immunosorbent assay (ELISA) and activated partial thromboplastin time (aPTT) assay in in vitro testing. Further characterization suggested ar3 sequences increased HC secretion, rather than promoting HC synthesis. Intravenous delivery of AAV8-HC(HL)+AAV8-LC or AAV8-HC(745)+AAV8-LC achieved phenotypic correction in hemophilia A mice. Mice receiving AAV8-HC(HL)+AAV8-LC achieved three- to fourfold higher HC expression than AAV8-HC(745)+AAV8-LC, consistent with the FVIII functional assays. HC(HL) should be substituted for HC(745) in a dual AAV vector strategy due to its enhanced expression.


Assuntos
Fator VIII/metabolismo , Terapia Genética , Hemofilia A/genética , Hemofilia A/terapia , Ácidos/química , Adenoviridae/genética , Animais , Antígenos/imunologia , Linhagem Celular , Fator VIII/química , Fator VIII/genética , Regulação da Expressão Gênica , Hemofilia A/imunologia , Hemofilia A/metabolismo , Humanos , Camundongos , Camundongos Knockout , Mutação/genética , Fenótipo
7.
Hum Gene Ther ; 17(4): 427-39, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16610930

RESUMO

We reported total correction of blood coagulation plasma factor VIII (FVIII) activity, using adeno-associated virus serotype 8 (AAV8) vectors for liver-specific gene transfer in hemophilia A mice. We now show, irrespective of immunosuppression or route of administration, total long-term correction of hemophilia A mice with pseudotyped AAV8 and AAV9 vectors. We delivered two FVIII vectors, one expressing canine heavy chain and the other expressing canine light chain. Interestingly, when these vectors were given by hepatic portal vein to hemophilia A dogs, only modest FVIII levels were seen despite the species-specific transgene. No dogs treated developed FVIII inhibitors. However, of three dogs treated with AAV8 vector, the single male, given 1.25 x 10(13) genome copies per vector per kilogram (GC/vector/kg), maintained a level of >4.5% for more than 2 years. In contrast, the two female dogs expressed only 2% FVIII activity despite receiving higher doses of 1.52 x 10(13) and 3 x 10(13) GC/vector/kg, respectively. On the other hand, a male dog treated with AAV9 vector at a low dose (6 x 10(12) GC/vector/kg) maintained FVIII levels of 2-2.5% of normal without bleeding for 200 days (observation ongoing). Although hemophilia A mice were not predictive of vector efficacy in dogs, the two treated male dogs became symptom-free for long periods. Even so, translation of these robust vectors either in appropriate large animals or human beings remains challenging.


Assuntos
Dependovirus , Fator VIII/genética , Terapia Genética , Vetores Genéticos , Hemofilia A/terapia , Animais , Ciclofosfamida/administração & dosagem , Cães , Fator VIII/uso terapêutico , Técnicas de Transferência de Genes , Hemofilia A/genética , Hemofilia A/imunologia , Humanos , Terapia de Imunossupressão , Imunossupressores/administração & dosagem , Fígado/irrigação sanguínea , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Veia Porta , Fatores de Tempo , Transgenes
8.
J Clin Invest ; 126(2): 405-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26829621

RESUMO

This position statement originated from a working group meeting convened on April 15, 2015, by the NHLBI and incorporates follow-up contributions by the participants as well as other thought leaders subsequently consulted, who together represent research fields relevant to all branches of the NIH. The group was deliberately composed not only of individuals with a current research emphasis in the glycosciences, but also of many experts from other fields, who evinced a strong interest in being involved in the discussions. The original goal was to discuss the value of creating centers of excellence for training the next generation of biomedical investigators in the glycosciences. A broader theme that emerged was the urgent need to bring the glycosciences back into the mainstream of biology by integrating relevant education into the curricula of medical, graduate, and postgraduate training programs, thus generating a critical sustainable workforce that can advance the much-needed translation of glycosciences into a more complete understanding of biology and the enhanced practice of medicine.


Assuntos
Pesquisa Biomédica/educação , Educação Profissionalizante , Glicômica/educação , Animais , Humanos
9.
Hum Gene Ther ; 14(2): 143-52, 2003 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-12614565

RESUMO

Hemophilia A is a sex-linked disorder that results from a deficiency of functional factor VIII and is currently treated by protein replacement therapies. Within the past decade, gene therapy efforts have come to the forefront of novel therapeutics. In this work, a dual-vector approach was employed in which recombinant adeno-associated viral (rAAV) vectors expressing the heavy and light chains of the murine factor VIII gene were delivered either intramuscularly or intravenously to a mouse model of hemophilia A. From in vitro work, it was determined that coinfection with both vectors is required as heterodimerization of the heavy and light chains occurs intracellularly. In vivo, therapeutic levels of factor VIII expression were achieved throughout the duration of the study (22 weeks). Intravenous and intramuscular delivery resulted in a maximal average expression of 31.4 +/- 6.4 and 29 +/- 6.5% of normal murine factor VIII levels, respectively. Western blots of cryoprecipitate as well as immunostaining of injection sites with an anti-murine factor VIII light chain antibody also confirmed the expression of factor VIII. Because the murine form of the gene was used in the mouse model, less than 1 Bethesda unit of inhibitors was noted. This work demonstrates the feasibility of using rAAV vectors for the long-term treatment of hemophilia A.


Assuntos
Fator VIII/genética , Terapia Genética , Vetores Genéticos , Hemofilia A/terapia , Animais , Dependovirus , Modelos Animais de Doenças , Fator VIII/metabolismo , Células HeLa , Humanos , Camundongos , Camundongos Knockout
11.
J Control Release ; 132(3): 252-9, 2008 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-18634839

RESUMO

Effective oral delivery of a non-viral gene carrier would represent a novel and attractive strategy for therapeutic gene transfer. To evaluate the potential of this approach, we studied the oral gene delivery efficacy of DNA polyplexes composed of chitosan and Factor VIII DNA. Transgene DNA was detected in both local and systemic tissues following oral administration of the chitosan nanoparticles to hemophilia A mice. Functional factor VIII protein was detected in plasma by chromogenic and thrombin generation assays, reaching a peak level of 2-4% FVIII at day 22 after delivery. In addition, a bleeding challenge one month after DNA administration resulted in phenotypic correction in 13/20 mice given 250-600 microg of FVIII DNA in chitosan nanoparticles, compared to 1/13 mice given naked FVIII DNA and 0/6 untreated mice. While further optimization would be required to render this type of delivery system practical for hemophilia A gene therapy, the findings suggest the feasibility of oral, non-viral delivery for gene medicine applications.


Assuntos
Quitosana/química , DNA/metabolismo , Fator VIII/biossíntese , Terapia Genética/métodos , Hemofilia A/terapia , Nanopartículas , Transfecção , Administração Oral , Animais , DNA/administração & dosagem , DNA/química , Modelos Animais de Doenças , Cães , Fator VIII/genética , Estudos de Viabilidade , Hemofilia A/sangue , Hemofilia A/genética , Hemostasia , Humanos , Camundongos , Camundongos Knockout , Trombina/metabolismo , Fatores de Tempo , Distribuição Tecidual
12.
Hum Gene Ther ; 19(6): 648-54, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18500941

RESUMO

Hemophilia A is caused by a deficiency in the factor VIII (FVIII) gene. Constrained by limited packaging capacity, even the 4.3-kb B domain-deleted FVIII remained a challenge for delivery by a single adeno-associated viral (AAV) vector. Studies have shown that up to a 6.6-kb vector sequence may be packaged into AAV virions, which suggested an alternative strategy for hemophilia A gene therapy. To explore the usefulness of AAV vectors carrying an oversized FVIII gene, we constructed the AAV-FVIII vector under the control of a beta-actin promoter with a cytomegalovirus enhancer (CB) and a bovine growth hormone (bGH) poly(A) sequence. The CB promoter plus bGH signal was shown to be 3- to 5-fold more potent than the mini-transthyretin (TTR) promoter with a synthetic poly(A) sequence for directing FVIII expression in the liver. Despite the 5.75-kb genome size of pAAV-CB-FVIII, sufficient AAV vectors were produced for in vivo testing. Approximately 3- to 5-fold more FVIII secretion was observed in animals receiving AAV-CB-FVIII vectors than in those receiving standard-sized AAV-TTR-FVIII vectors. Both the activated partial thromboplastin time assay and the whole blood thromboelastographic analysis confirmed that AAV-FVIII vectors fully corrected the bleeding phenotype of hemophilia mice. These results suggest that AAV vectors with an oversized genome should be useful for not only hemophilia A gene therapy but also other diseases with large cDNA such as muscular dystrophy and cystic fibrosis.


Assuntos
Dependovirus , Fator VIII/genética , Terapia Genética/métodos , Vetores Genéticos , Hemofilia A/terapia , Animais , Linhagem Celular , Humanos , Camundongos , Camundongos Knockout
13.
Mol Ther ; 15(10): 1856-62, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17653101

RESUMO

Coagulation factor VIII (FVIII) is secreted as a heterodimer consisting of a heavy chain (HC) and a light chain (LC), which can be expressed independently and reassociate with recovery of biological activity. Because of the size limitation of adeno-associated virus (AAV) vectors, a strategy for delivering the HC and LC separately has been developed. However, the FVIII HC is secreted 10-100-fold less efficiently than the LC. In this study, we demonstrated that the F309S mutation and enhanced B-domain glycosylations alone are not sufficient to improve FVIII HC secretion, which suggested a role of the FVIII LC in regulating HC secretion. To characterize this role of the FVIII LC, we compared FVIII HC secretion with and without the LC via post-translational protein trans-splicing. As demonstrated in vitro, ligation of the LC to the HC significantly increased HC secretion. Such HC secretion increases were also confirmed in vivo by hydrodynamic injection of FVIII intein plasmids into hemophilia A mice. Moreover, similar enhancement of HC secretion can also be observed when the LC is supplied in trans, which is probably due to the spontaneous association of the HC and the LC in the secretion pathway. In sum, enhancing the secretion of the FVIII HC polypeptide may require the proper association of the FVIII LC polypeptide in cis or in trans. These results may be helpful in designing new strategies to improve FVIII gene delivery.


Assuntos
Fator VIII/genética , Animais , Western Blotting , Linhagem Celular , Fator VIII/química , Fator VIII/metabolismo , Humanos , Camundongos , Camundongos Knockout , Plasmídeos , Processamento de Proteína Pós-Traducional
14.
J Biol Chem ; 280(18): 17593-600, 2005 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-15728582

RESUMO

Factor VIII is a multidomain protein composed of A1, A2, B, A3, C1, and C2 domains. Deficiency or dysfunction of factor VIII causes hemophilia A, a bleeding disorder. Administration of exogenous recombinant factor VIII as a replacement leads to development of inhibitory antibodies against factor VIII in 15-30% of hemophilia A patients. Hence, less immunogenic preparations of factor VIII are highly desirable. Inhibitory antibodies against factor VIII are mainly directed against immunodominant epitopes in C2, A3, and A2 domains. Further, several universal epitopes for CD4+ T-cells have been identified within the C2 domain. The C2 domain is also known to interact specifically with phosphatidylserine-rich lipid vesicles. Here, we have investigated the hypothesis that complexation of O-phospho-l-serine, the head group of phosphatidylserine, with the C2 domain can reduce the overall immunogenicity of factor VIII. The biophysical (circular dichroism and fluorescence) and biochemical studies (ELISA and size exclusion chromatography) showed that O-phospho-l-serine binds to the phospholipid-binding region in the C2 domain, and this interaction causes subtle changes in the tertiary structure of the protein. O-Phospho-l-serine also prevented aggregation of the protein under thermal stress. The immunogenicity of the factor VIII-O-phospho-l-serine complex was evaluated in hemophilia A mice. The total and inhibitory antibody titers were lower for factor VIII-O-phospho-l-serine complex compared with factor VIII alone. Moreover, factor VIII administered as a complex with O-phospho-l-serine retained in vivo activity in hemophilia A mice. Our results suggest that factor VIII-O-phospho-l-serine complex may be beneficial to increase the physical stability and reduce immunogenicity of recombinant factor VIII preparations.


Assuntos
Fator VIII/administração & dosagem , Fator VIII/imunologia , Hemofilia A/tratamento farmacológico , Hemofilia A/imunologia , Isoanticorpos/biossíntese , Fosfosserina/análogos & derivados , Fosfosserina/administração & dosagem , Animais , Fator VIII/uso terapêutico , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
15.
Proc Natl Acad Sci U S A ; 102(17): 6080-5, 2005 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-15837921

RESUMO

Hemophilia A (HA) is a bleeding disorder caused by factor VIII (FVIII) deficiency. FVIII replacement therapy can reduce bleeding but is expensive, inconvenient, and complicated by development of antibodies that inhibit FVIII activity in 30% of patients. Neonatal hepatic gene therapy could result in continuous secretion of FVIII into blood and might reduce immunological responses. Newborn HA mice and dogs that were injected i.v. with a retroviral vector (RV) expressing canine B domain-deleted FVIII (cFVIII) achieved plasma cFVIII activity that was 139 +/- 22% and 116 +/- 5% of values found in normal dogs, respectively, which was stable for 1.5 yr. Coagulation tests were normalized, no bleeding had occurred, and no inhibitors were detected. This is a demonstration of long-term fully therapeutic gene therapy for HA in a large animal model. Desmopressin (DDAVP; 1-deamino-[d-Arg(8)]vasopressin) is a drug that increases FVIII activity by inducing release of FVIII complexed with von Willebrand factor from endothelial cells. It has been unclear, however, if the FVIII is synthesized by endothelial cells or is taken up from blood. Because the plasma cFVIII in these RV-treated dogs derives primarily from transduced hepatocytes, they provided a unique opportunity to study the biology of the DDAVP response. Here we show that DDAVP did not increase plasma cFVIII levels in the RV-treated dogs, although von Willebrand factor was increased appropriately. This result suggests that the increase in FVIII in normal dogs after DDAVP is due to release of FVIII synthesized by endothelial cells.


Assuntos
Desamino Arginina Vasopressina/uso terapêutico , Fator VIII/genética , Hemofilia A/genética , Fígado/fisiologia , Animais , Animais Recém-Nascidos , Clonagem Molecular , DNA Complementar/genética , Doenças do Cão/genética , Cães , Terapia Genética , Vetores Genéticos , Hemofilia A/veterinária , Humanos , Fígado/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Retroviridae , Fator de von Willebrand
16.
Blood ; 103(4): 1253-60, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14551134

RESUMO

Despite the popularity of adeno-associated virus 2 (AAV2) as a vehicle for gene transfer, its efficacy for liver-directed gene therapy in hemophilia A or B has been suboptimal. Here we evaluated AAV serotypes 2, 5, 7, and 8 in gene therapy of factor VIII (FVIII) deficiency in a hemophilia A mouse model and found that AAV8 was superior to the other 3 serotypes. We expressed canine B domain-deleted FVIII cDNA either in a single vector or in 2 separate AAV vectors containing the heavy- and light-chain cDNAs. We also evaluated AAV8 against AAV2 in intraportal and tail vein injections. AAV8 gave 100% correction of plasma FVIII activity irrespective of the vector type or route of administration.


Assuntos
Adenoviridae/genética , Fator VIII/genética , Terapia Genética/métodos , Hemofilia A/terapia , Adenoviridae/classificação , Animais , Southern Blotting , DNA Recombinante/farmacocinética , Cães , Feminino , Vetores Genéticos , Hemofilia A/genética , Fígado , Masculino , Camundongos , Camundongos Mutantes , Veia Porta , Cauda/irrigação sanguínea
17.
Mol Ther ; 10(1): 117-26, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15233948

RESUMO

The development of antibodies to a previously unexpressed protein product may limit the success of human gene therapy approaches. We inserted B-domain-deleted factor VIII (FVIII) cDNA of human, canine, or murine origin into the multiple cloning site of a liver-specific vector, pBS-HCRHPI-A, to yield plasmids pBS-HCRHPI-FVIIIA, pBS-HCRHPI-cFVIIIA, and pBS-HCRHPI-mFVIIIA, respectively. Fifty micrograms of each plasmid in 2 ml of solution was rapidly injected into the tail vein of three groups of hemophilia A mice. Factor VIII levels ranging from 3 to 12 IU/ml were obtained from all three groups (normal is 1 IU/ml in human plasma) 3 days after treatment. These initial very high levels of functional human, canine, or murine factor VIII, however, fell gradually to undetectable levels within 2-3 weeks, and their disappearance correlated with the generation of high-titer, inhibitory anti-FVIII antibodies. Notably, this immune response occurred independent of the species of origin of the exogenous factor VIII. Antibody titers to factor VIII were detected beginning at 2 weeks, reached a plateau and remained at high levels for over 6 months. The majority of anti-hFVIII IgG was IgG1 isotype specific, suggesting a humoral response mediated by Th2-induced signals. Consistent with this idea, in a separate group of mice treated with pBS-HCRHPI-FVIIIA, transient immunosuppression by cyclophosphamide significantly delayed (5/6) or abolished (1/6) inhibitory antibody formation against the transgene.


Assuntos
Fator VIII/genética , Fator VIII/imunologia , Terapia Genética , Hemofilia A/imunologia , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Animais , Formação de Anticorpos/efeitos dos fármacos , Ciclofosfamida/farmacologia , DNA/administração & dosagem , Cães , Fator VIII/metabolismo , Vetores Genéticos/análise , Vetores Genéticos/genética , Hemofilia A/terapia , Humanos , Imunoglobulina G/análise , Imunoglobulina G/metabolismo , Fígado/química , Fígado/metabolismo , Camundongos , Fragmentos de Peptídeos/metabolismo , Plasmídeos/análise , Plasmídeos/genética , Transgenes
18.
Blood ; 102(12): 3919-26, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12893764

RESUMO

Using separate adeno-associated viral 2 (AAV2) vectors to deliver the heavy and light chains of factor VIII (FVIII) we have overcome the packaging limitations of AAV, achieving phenotypic correction of hemophilia A in mice. AAV vectors were constructed that use a liver-specific promoter and the cDNA sequences of either the human or canine heavy and light chains of FVIII. After intraportal vein injection of these vectors in hemophilia-A mice, therapeutic to superphysiologic levels of active FVIII were achieved in plasma in a dose-dependent manner. Phenotypic correction of the bleeding diathesis was demonstrated by survival of all treated mice after tail clipping. Biochemical analysis demonstrated lower levels of heavy-chain (25- to 100-fold) compared with light-chain protein in the plasma of treated animals. Differences in gene transfer and transcription did not account for the differences in protein expression. We hypothesize that improvements in FVIII activity could be achieved by improvements in FVIII heavy-chain expression. This work demonstrates that cotransduction of liver with AAV vectors expressing the heavy and light chains of FVIII corrects hemophilia A in vivo, providing an alternative approach to the use of a single vector. This strategy may potentially be useful for other large therapeutic proteins that contain functionally distinct domains.


Assuntos
Fator VIII/administração & dosagem , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Hemofilia A/terapia , Subunidades Proteicas/administração & dosagem , Adenoviridae/genética , Animais , Modelos Animais de Doenças , Cães , Fator VIII/análise , Fator VIII/genética , Hemorragia/prevenção & controle , Humanos , Fígado/metabolismo , Camundongos , Camundongos Knockout , Fenótipo , Veia Porta , Regiões Promotoras Genéticas , Subunidades Proteicas/sangue , Subunidades Proteicas/genética , Transgenes , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA