Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Mol Ther ; 22(9): 1614-24, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25034357

RESUMO

The development of robust nonviral vectors could facilitate clinical gene therapy applications and may overcome some of the immune complications of viral vectors. Nevertheless, most nonviral gene deliver approaches typically yield only transient and/or low gene expression. To address these caveats, we have explored piggyBac transposons to correct hemophilia B by liver-directed factor IX (FIX) gene therapy in hemophilic mice. To achieve this, we combined the use of: (i) a hyperactive codon-optimized piggyBac transposase, (ii) a computationally enhanced liver-specific promoter, (iii) a hyperfunctional codon-optimized FIX transgene (FIX R338L Padua), and (iv) a modification of the transposon terminal repeats. This combination strategy resulted in a robust 400-fold improvement in vector performance in hepatocytes, yielding stable supraphysiologic human FIX activity (>1 year). Liver-specific expression resulted in the induction of FIX-specific immune tolerance. Remarkably, only very low transposon/transposase doses were required to cure the bleeding diathesis. Similarly, PB transposons could be used to express supraphysiologic factor VIII levels using low transposon/transposase doses. PB transposition did not induce tumors in a sensitive hepatocellular carcinoma-prone mouse model. These results underscore the potency and relative safety of the latest generation PB transposons, which constitutes a versatile platform for stable and robust secretion of therapeutic proteins.


Assuntos
Elementos de DNA Transponíveis , Fator IX/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Hemofilia B/terapia , Hepatócitos/metabolismo , Animais , Modelos Animais de Doenças , Vetores Genéticos/uso terapêutico , Hemofilia B/imunologia , Hepatócitos/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Transposases/genética , Transposases/metabolismo
2.
Blood ; 117(3): 798-807, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-21041718

RESUMO

Gene therapy for hemophilia A would be facilitated by development of smaller expression cassettes encoding factor VIII (FVIII), which demonstrate improved biosynthesis and/or enhanced biologic properties. B domain deleted (BDD) FVIII retains full procoagulant function and is expressed at higher levels than wild-type FVIII. However, a partial BDD FVIII, leaving an N-terminal 226 amino acid stretch (N6), increases in vitro secretion of FVIII tenfold compared with BDD-FVIII. In this study, we tested various BDD constructs in the context of either wild-type or codon-optimized cDNA sequences expressed under control of the strong, ubiquitous Spleen Focus Forming Virus promoter within a self-inactivating HIV-based lentiviral vector. Transduced 293T cells in vitro demonstrated detectable FVIII activity. Hemophilic mice treated with lentiviral vectors showed expression of FVIII activity and phenotypic correction sustained over 250 days. Importantly, codon-optimized constructs achieved an unprecedented 29- to 44-fold increase in expression, yielding more than 200% normal human FVIII levels. Addition of B domain sequences to BDD-FVIII did not significantly increase in vivo expression. These significant findings demonstrate that shorter FVIII constructs that can be more easily accommodated in viral vectors can result in increased therapeutic efficacy and may deliver effective gene therapy for hemophilia A.


Assuntos
Códon/genética , Fator VIII/genética , Terapia Genética/métodos , Hemofilia A/terapia , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Ensaio de Imunoadsorção Enzimática , Fator VIII/metabolismo , Feminino , Expressão Gênica , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Células HEK293 , Hemofilia A/sangue , Hemofilia A/genética , Humanos , Injeções Intravenosas , Lentivirus/genética , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Dados de Sequência Molecular , Mutação , Regiões Promotoras Genéticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Vírus Formadores de Foco no Baço/genética
3.
J Pathol ; 223(5): 593-603, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21341275

RESUMO

Mesoangioblasts (MABs) are a subset of muscle-derived pericytes able to restore dystrophic phenotype in mice and dogs. However, their lifespan is limited and they undergo senescence after 25-30 population doublings. Recently, induced pluripotent stem cells (iPSCs) generated from reprogrammed fibroblasts have been demonstrated to have in vitro and in vivo myogenic potential when sorted for the SM/C-2.6 antigen. Furthermore, chimeric mice from mdx-iPSCs (DYS-HAC) cells showed tissue-specific expression of dystrophin. Nevertheless, myogenic differentiation protocols and the potential of iPSCs generated from different cell sources still present unanswered questions. Here we show that iPSCs generated from prospectively sorted MABs (MAB-iPSCs) are pluripotent as fibroblast-derived iPSCs (f-iPSCs). However, both teratoma formation and genetic cell manipulation assays identify a durable epigenetic memory in MAB-iPSCs, resulting in stronger myogenic commitment. Striated muscle tissue accounts for up to 70% of MAB-iPSC teratomas. Moreover, transfection with Pax3 and Pax7 induces a more robust myogenic differentiation in MAB-iPSCs than in f-iPSCs. A larger amount of CD56(+) progenitors can be sorted from the MAB-iPSCs differentiating pool and, after transplantation into αsg-KO mice, can efficiently participate to skeletal muscle regeneration and restore αsg expression. Our data strongly suggest that iPSCs are a heterogeneous population and, when generated from myogenic adult stem cells, they exhibit a stronger commitment, paving the way for creating custom-made cell protocols for muscular dystrophies.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Distrofia Muscular Animal/terapia , Pericitos/citologia , Animais , Animais Recém-Nascidos , Antígeno CD56/análise , Diferenciação Celular , Proliferação de Células , Elementos de DNA Transponíveis , Modelos Animais de Doenças , Epigênese Genética , Células-Tronco Pluripotentes Induzidas/transplante , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/fisiologia , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patologia , Regeneração , Sarcoglicanas/deficiência , Sarcoglicanas/metabolismo , Transfecção
4.
Blood ; 114(8): 1461-8, 2009 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-19471016

RESUMO

Effective gene therapy requires robust delivery of the desired genes into the relevant target cells, long-term gene expression, and minimal risks of secondary effects. The development of efficient and safe nonviral vectors would greatly facilitate clinical gene therapy studies. However, nonviral gene transfer approaches typically result in only limited stable gene transfer efficiencies in most primary cells. The use of nonviral gene delivery approaches in conjunction with the latest generation transposon technology based on Sleeping Beauty (SB) or piggyBac transposons may potentially overcome some of these limitations. In particular, a large-scale genetic screen in mammalian cells yielded a novel hyperactive SB transposase, resulting in robust and stable gene marking in vivo after hematopoietic reconstitution with CD34(+) hematopoietic stem/progenitor cells in mouse models. Moreover, the first-in-man clinical trial has recently been approved to use redirected T cells engineered with SB for gene therapy of B-cell lymphoma. Finally, induced pluripotent stem cells could be generated after genetic reprogramming with piggyBac transposons encoding reprogramming factors. These recent developments underscore the emerging potential of transposons in gene therapy applications and induced pluripotent stem generation for regenerative medicine.


Assuntos
Elementos de DNA Transponíveis/fisiologia , Técnicas de Transferência de Genes , Terapia Genética/tendências , Células-Tronco Pluripotentes/fisiologia , Animais , Técnicas de Transferência de Genes/tendências , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Humanos , Camundongos , Modelos Biológicos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Ativação Transcricional/fisiologia
5.
Stem Cells ; 28(10): 1760-71, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20715185

RESUMO

Adult stem cells and induced pluripotent stem cells (iPS) hold great promise for regenerative medicine. The development of robust nonviral approaches for stem cell gene transfer would facilitate functional studies and potential clinical applications. We have previously generated hyperactive transposases derived from Sleeping Beauty, using an in vitro molecular evolution and selection paradigm. We now demonstrate that these hyperactive transposases resulted in superior gene transfer efficiencies and expression in mesenchymal and muscle stem/progenitor cells, consistent with higher expression levels of therapeutically relevant proteins including coagulation factor IX. Their differentiation potential and karyotype was not affected. Moreover, stable transposition could also be achieved in iPS, which retained their ability to differentiate along neuronal, cardiac, and hepatic lineages without causing cytogenetic abnormalities. Most importantly, transposon-mediated delivery of the myogenic PAX3 transcription factor into iPS coaxed their differentiation into MYOD(+) myogenic progenitors and multinucleated myofibers, suggesting that PAX3 may serve as a myogenic "molecular switch" in iPS. Hence, this hyperactive transposon system represents an attractive nonviral gene transfer platform with broad implications for regenerative medicine, cell and gene therapy.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Diferenciação Celular/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Retroelementos/fisiologia , Animais , Diferenciação Celular/genética , Linhagem Celular , Humanos , Camundongos , Células Musculares/citologia , Células Musculares/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Retroelementos/genética , Transposases/metabolismo
6.
Mol Ther ; 18(3): 477-90, 2010 03.
Artigo em Inglês | MEDLINE | ID: mdl-20087315

RESUMO

Lentiviral vectors (LVs) have emerged as potent and versatile vectors for ex vivo or in vivo gene transfer into dividing and nondividing cells. Robust phenotypic correction of diseases in mouse models has been achieved paving the way toward the first clinical trials. LVs can deliver genes ex vivo into bona fide stem cells, particularly hematopoietic stem cells, allowing for stable transgene expression upon hematopoietic reconstitution. They are also useful to generate induced pluripotent stem cells. LVs can be pseudotyped with distinct viral envelopes that influence vector tropism and transduction efficiency. Targetable LVs can be generated by incorporating specific ligands or antibodies into the vector envelope. Immune responses toward the transgene products and transduced cells can be repressed using microRNA-regulated vectors. Though there are safety concerns regarding insertional mutagenesis, their integration profile seems more favorable than that of gamma-retroviral vectors (gamma-RVs). Moreover, it is possible to minimize this risk by modifying the vector design or by employing integration-deficient LVs. In conjunction with zinc-finger nuclease technology, LVs allow for site-specific gene correction or addition in predefined chromosomal loci. These recent advances underscore the improved safety and efficacy of LVs with important implications for clinical trials.


Assuntos
Terapia Genética/métodos , Vetores Genéticos , Lentivirus/genética , Animais , Técnicas de Transferência de Genes , Células-Tronco Hematopoéticas/citologia , Humanos , Ligantes , Modelos Genéticos , Mutagênese , Fenótipo , Células-Tronco Pluripotentes/citologia , Retroviridae/genética , Risco , Transgenes
7.
Mol Ther ; 18(6): 1200-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20372108

RESUMO

Transposon-based gene vectors have become indispensable tools in vertebrate genetics for applications ranging from insertional mutagenesis and transgenesis in model species to gene therapy in humans. The transposon toolkit is expanding, but a careful, side-by-side characterization of the diverse transposon systems has been lacking. Here we compared the Sleeping Beauty (SB), piggyBac (PB), and Tol2 transposons with respect to overall activity, overproduction inhibition (OPI), target site selection, transgene copy number as well as long-term expression in human cells. SB was the most efficient system under conditions where the availability of the transposon DNA is limiting the transposition reaction including hard-to-transfect hematopoietic stem/progenitor cells (HSCs), and the most sensitive to OPI, underpinning the need for careful optimization of the transposon components. SB and PB were about equally active, and both more efficient than Tol2, under nonrestrictive conditions. All three systems provided long-term transgene expression in human cells with minimal signs of silencing. Indeed, mapping of Tol2 insertion sites revealed significant underrepresentation within chromosomal regions with H3K27me3 histone marks typically associated with transcriptionally repressed heterochromatin. SB, Tol2, and PB constitute complementary research tools for gene transfer in mammalian cells with important implications for fundamental and translational research.


Assuntos
Elementos de DNA Transponíveis , Vetores Genéticos , Células HeLa , Humanos , Transgenes
8.
Curr Opin Hematol ; 17(5): 387-92, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20657281

RESUMO

PURPOSE OF REVIEW: Hemophilia A and B are attractive target diseases for gene therapy, as stable expression of coagulation factor VIII and IX may correct the bleeding diathesis. This review focuses on the recent progress in preclinical and clinical studies in gene therapy for hemophilia A and B. RECENT FINDINGS: Hepatic gene delivery using vectors derived from adeno-associated virus (AAV) resulted in therapeutic but transient functional clotting factor IX (FIX) expression levels in severe hemophilia B patients. Although T-cell-mediated immune responses eliminated the transduced hepatocytes, transient immunosuppression may potentially overcome this limitation. Alternatively, vectors are being developed that result in higher FIX expression levels at lower vector doses. Lentiviral vectors are being explored for in-vivo hepatic gene delivery and for ex-vivo transduction of hematopoietic stem cells. This resulted in stable correction of the bleeding diathesis in hemophilic mice. Finally, nonviral vectors derived from transposons result in sustained clotting-factor expression in rodent models. Translational studies in large animal models are required to move these new approaches forward into the clinic. SUMMARY: New insights from clinical trials and advances in preclinical studies may ultimately pave the way toward a cure in patients suffering from hemophilia.


Assuntos
Terapia Genética/métodos , Hemofilia A/terapia , Hemofilia B/terapia , Animais , Modelos Animais de Doenças , Vetores Genéticos , Hepatócitos , Humanos , Camundongos , Linfócitos T Reguladores
9.
Methods ; 49(3): 287-97, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19615447

RESUMO

Transposable elements can be considered as natural, non-viral gene delivery vehicles capable of efficient genomic insertion. The plasmid-based transposon system of Sleeping Beauty (SB) combines the advantages of viruses and naked DNA molecules. In contrast to plasmid vectors, transposons integrate through a precise, recombinase-mediated mechanism into chromosomes, providing long-term expression of the gene of interest in cells. The advantages of transposons in comparison to viral systems include their simplicity and improved safety/toxicity profiles. In addition, the hyperactive SB100X is the first plasmid-based delivery system that overcomes the efficacy of non-viral delivery. The transposon delivery system consists of the transposase and the integration cassette, recognized by the transposase. The plasmid-based transposon delivery system can be combined with any non-viral delivery method. Here we provide two detailed protocols to apply SB-mediated, non-viral gene transfer in cultured cells. In our first example, we use a lipid-based delivery method in combination with the transposon-based integration system in an easy-to-transfect (HeLa) cell line. Second, we show how to achieve 40-50% stable expression of a transgene in clinically relevant, hard-to-transfect cells (hematopoetic stem cells, HSCs) by nucleofection. The given protocols are adaptable to any vertebrate cells in culture.


Assuntos
Elementos de DNA Transponíveis , Técnicas de Transferência de Genes , Vetores Genéticos , Células HeLa , Células-Tronco Hematopoéticas , Humanos
10.
Arterioscler Thromb Vasc Biol ; 28(9): 1621-6, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18556568

RESUMO

OBJECTIVE: Gene therapy for severe von Willebrand disease (vWD) seems an interesting treatment alternative with long-term therapeutic potential. We investigated the feasibility of targeting the liver for ectopic expression of physiologically active von Willebrand factor (vWF). METHODS AND RESULTS: The capacity of transgene-encoded murine vWF to restore vWF function was studied in a mouse model of severe vWD after liver-specific gene transfer by hydrodynamic injection. By using a hepatocyte-specific alpha1 antitrypsin promoter, a considerably higher and longer-lasting vWF expression was obtained when compared with a cytomegalovirus promoter, reaching maximum vWF plasma levels that are 10+/-1 times higher than the wild-type level. Liver-expressed vWF showed the full range of multimers, including the high molecular weight multimers, and restored factor VIII plasma levels, consistent with correction of the bleeding time 3 but not 7 days after gene transfer. Importantly, transgene encoded plasma vWF restored proper platelet adhesion and aggregation in a FeCl(3) induced thrombosis model. CONCLUSIONS: High ectopic expression of transgene encoded plasma vWF can be obtained after gene transfer to the liver. Liver-expressed vWF was fully multimerized and able to restore proper platelet plug formation in severe vWD. The liver therefore seems an attractive target for gene therapy for severe vWD.


Assuntos
Terapia Genética/métodos , Fígado/metabolismo , Trombose/terapia , Doenças de von Willebrand/terapia , Fator de von Willebrand/metabolismo , Animais , Tempo de Sangramento , Cloretos , Citomegalovirus/genética , Modelos Animais de Doenças , Fator VIII/metabolismo , Estudos de Viabilidade , Compostos Férricos , Técnicas de Transferência de Genes , Humanos , Camundongos , Camundongos Knockout , Adesividade Plaquetária , Agregação Plaquetária , Regiões Promotoras Genéticas , Índice de Gravidade de Doença , Trombose/sangue , Trombose/induzido quimicamente , Trombose/genética , Fatores de Tempo , alfa 1-Antitripsina/genética , Doenças de von Willebrand/sangue , Doenças de von Willebrand/genética , Fator de von Willebrand/genética
11.
Nat Biotechnol ; 21(8): 885-90, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12833071

RESUMO

Hepatitis B virus envelope L particles form hollow nanoparticles displaying a peptide that is indispensable for liver-specific infection by hepatitis B virus in humans. Here we demonstrate the use of L particles for the efficient and specific transfer of a gene or drug into human hepatocytes both in culture and in a mouse xenograft model. In this model, intravenous injection of L particles carrying the gene for green fluorescent protein (GFP) or a fluorescent dye resulted in observable fluorescence only in human hepatocellular carcinomas but not in other human carcinomas or in mouse tissues. When the gene encoding human clotting factor IX was transferred into the xenograft model using L particles, factor IX was produced at levels relevant to the treatment of hemophilia B. The yeast-derived L particle is free of viral genomes, highly specific to human liver cells and able to accommodate drugs as well as genes. These advantages should facilitate targeted delivery of genes and drugs to the human liver.


Assuntos
DNA/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Marcação de Genes/métodos , Hepatócitos/metabolismo , Nanotecnologia/métodos , Neoplasias/metabolismo , Transfecção/métodos , Proteínas do Envelope Viral/farmacocinética , Animais , Linhagem Celular Tumoral , Eletroporação/métodos , Estudos de Viabilidade , Terapia Genética/métodos , Humanos , Masculino , Camundongos , Microesferas , Dados de Sequência Molecular , Nanotecnologia/instrumentação , Tamanho da Partícula
12.
IDrugs ; 8(10): 818-21, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16254801

RESUMO

Gene therapy is a promising biomedical discipline that could potentially lead to new treatments and perhaps long-term curative effects for a plethora of diseases including hereditary disorders, cardiovascular and neurological diseases, cancer, diabetes and even infectious or autoimmune diseases. These diseases affect millions of people worldwide and the development of effective and safe gene-based drugs obviously represent a tremendous market potential. Convincing evidence continues to emerge from clinical trials demonstrating that gene therapy can be effective in patients suffering from a limited number of different diseases. Nevertheless, as with any emerging new biomedical discipline, gene therapy has also faced a number of setbacks, and there have been concerns regarding the safety of some gene delivery approaches, however, these hurdles are not insurmountable. Gene transfer technologies are improving rapidly and have led to the development of new and more efficacious gene delivery approaches with fewer side effects. The success of gene therapy is still highly dependent upon the continuous development of improved gene delivery technologies, the progress of which should hopefully and ultimately cure diseases that are refractory to current treatment paradigms.


Assuntos
Técnicas de Transferência de Genes/tendências , Terapia Genética/normas , Terapia Genética/tendências , Terapia Genética/métodos , Humanos , Resultado do Tratamento
13.
Hum Gene Ther ; 14(3): 263-76, 2003 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-12639306

RESUMO

B lymphocytes are attractive targets for gene therapy of genetic diseases associated with B-cell dysfunction and for immunotherapy. Transduction of B lymphocytes was evaluated using green fluorescent protein (GFP)-encoding onco-retroviral and HIV-derived lentiviral vectors which were pseudotyped with ecotropic, amphotropic or vesicular stomatitis virus (VSV-G) envelopes. Transduction of mouse B lymphocytes activated with lipopolysaccharides (LPS) or by cross-linking CD40 in conjunction with interleukin-4 (IL-4) was significantly more efficient (p < 0.003) with ecotropic (11%) than with VSV-G pseudotyped onco-retroviral vectors (1%). Using high-titer cell-free ecotropic viral supernatant or by coculture with ecotropic onco-retroviral vector-producing cells, transduction efficiency increased significantly (p < 0.001) to approximately 50%, whereas transduction efficiency by coculture with VSV-G pseudotyped vector-producing cells remained low (< 2%). Similarly, transduction of mouse B lymphocytes was significantly more efficient (twofold, p < 0.01) with the ecotropic (7%) than with the VSV-G pseudotyped lentiviral vectors although gene transfer efficiency remained low because of dose-limiting toxicity of the concentrated vector preparations on the LPS-activated murine B cells. Consistent with murine B-cell transduction, human B cells activated with CD40L and IL-4 were also found to be relatively refractory to VSV-G pseudotyped onco-retroviral vectors (< 1%). However, higher transduction efficiencies could be achieved in activated primary human B lymphocytes using VSV-G pseudotyped lentiviral vectors instead (5%-6%). Contrary to the significant increase in mouse B-cell transduction efficiency with ecotropic vectors, the use of amphotropic onco-retroviral or lentiviral vectors did not increase transduction efficiency in primary human B cells. The present study shows that the transduction efficiency of onco-retroviral and lentiviral vectors in human and mouse B lymphocytes is pseudotype-dependent and challenges the widely held assumption that VSV-G pseudotyping facilitates gene transfer into all cell types.


Assuntos
Linfócitos B/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Lentivirus , Retroviridae , Animais , Vetores Genéticos/toxicidade , Humanos , Camundongos , Transdução Genética
14.
Curr Gene Ther ; 3(6): 501-15, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14683449

RESUMO

Extensive gene therapy studies in preclinical models and in clinical trials underscore the relative safety of onco-retroviral vectors. Up until recently, no adverse effects have been reported in nearly 2000 patients that were enrolled in gene therapy clinical trials involving onco-retroviral vectors. However, the main safety concern of using onco-retroviral vectors is related to the risk of malignant transformation following oncogene activation due to random onco-retroviral genomic integration. Based on primate studies, there is an apparent low risk of malignancy that is predominately associated with the occurrence of chronic retroviremia resulting from replication-competent retroviruses (RCR), particularly in immunosuppressed recipient hosts. However, in the latest packaging cell lines and vectors, the risk of RCR-generation has been drastically reduced, primarily by minimizing the homologous overlap between vector and helper sequences. Nevertheless, results from a recent preclinical study in mice and a clinical trial in patients suffering from SCID-X1 strongly suggest that onco-retroviral vectors devoid of RCR can contribute to lymphomagenesis by insertional activation of cellular oncogenes. The risk of inadvertent germline transmission of onco-retroviral vectors appears to be low, especially relative to the endogenous rate of germline insertion, which is known to occur naturally in the human population via transmission of endogenous retro-transposons. The strict dependency of onco-retroviral gene transfer on cell division is an important safety advantage that significantly limits the risks of horizontal transmission. Since improved onco-retroviral vectors or transduction protocols may result in an increased number of retroviral integrations per cell, this may concomitantly increase the risk of malignant transformation. The use of suicide genes, self-inactivating vectors and/or chromosomal insulators is, therefore, warranted to further enhance the safety features of onco-retroviral vectors. Detailed analyses of insertion sites combined with long-term clinical follow-up may contribute to a more accurate risk assessment.


Assuntos
Vetores Genéticos/efeitos adversos , Oncogenes , Retroviridae/genética , Gestão da Segurança , Animais , Linhagem Celular , Humanos , Camundongos , Montagem de Vírus
15.
Curr Gene Ther ; 3(6): 527-43, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14683450

RESUMO

Adenoviral vectors can efficiently transduce a broad variety of different cell types and have been used extensively in preclinical and clinical studies. However, early generation of adenoviral vectors retained residual adenoviral genes that contribute to inflammatory immune responses and toxicity. In addition, these vectors often result in transient expression of the potentially therapeutic transgene. Some clinical trials based on early generation adenoviral vectors have been discontinued because of acute inflammatory responses and toxicity and even one patient has died as a direct consequence of adenoviral toxicity. The latest generation of high-capacity adenoviral vectors is devoid of viral genes, and is having a significantly improved safety profile and yielding more prolonged transgene expression compared to early generation vectors. Nevertheless, transgene expression gradually declines even when high-capacity adenoviral vectors are used, possibly due to the gradual loss of vector genomes. Despite their improved safety, high-capacity adenoviral vectors can still trigger transient toxic effects in animals and patients. Restricting the tropism of adenoviral vectors by immunologic or genetic re-targeting may further improve their therapeutic window. The safety of adenoviral vectors has been improved further through the development of safer packaging systems that eliminate the homologous overlap between vector and helper sequences and therefore prevent formation of replication-competent adenoviruses (RCA). RCA could exacerbate inflammatory responses and act as a helper to rescue adenoviral vectors, potentially increasing the effective vector dose. Conditionally replicating adenoviruses (CRAds) have been developed for cancer gene therapy, which replicate selectively in some cancer cells. The use of CRAds in combination with chemotherapy yielded therapeutic effects in patients suffering from cancer but dose-limiting toxicity was apparent. Although there appears to be a very low theoretical risk of malignancy that is predominantly associated with the occurrence of E1-positive recombinants, no malignancies have been reported that were associated with adenoviral vectors. Nevertheless, integrating adenoviral vectors carry a greater malignancy risk due to their ability to integrate randomly into the target genomes.


Assuntos
Adenoviridae/genética , Vetores Genéticos/efeitos adversos , Gestão da Segurança , Adenoviridae/fisiologia , Animais , Feminino , Camundongos
16.
Curr Gene Ther ; 2(2): 195-209, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12109216

RESUMO

The bone marrow (BM) is composed of the non-adherent hematopoietic and adherent stromal cell compartment. This adherent BM stromal cell fraction contains pluripotent mesenchymal stem cells (MSCs) and differentiated mesenchymal BM stromal cells. The MSCs self-renew by proliferation while maintaining their stem-cell phenotype and give rise to the differentiated stromal cells which belong to the osteogenic, chondrogenic, adipogenic, myogenic and fibroblastic lineages. A more primitive adherent stem cell was recently identified, the multipotent adult progenitor cell (MAPC) or mesodermal progenitor cell, which co-purifies with MSCs. These MAPCs differentiate into MSCs, endothelial, epithelial and even hematopoietic cells. BM stroma cells, including the primitive pluripotent MSCs and MAPCs, are attractive targets for cell and gene therapy. The BM stromal cell population and its multipotent stem cells can be engineered to secrete a series of different proteins in vitro and in vivo that could potentially treat a variety of serum protein deficiencies and other genetic or acquired diseases, including bone, cartilage and BM stromal disorders or even cancer.


Assuntos
Células da Medula Óssea/citologia , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Células Estromais/citologia , Adenoviridae/genética , Animais , Adesão Celular , Diferenciação Celular , Divisão Celular , Células Cultivadas , Ensaios Clínicos como Assunto , Terapia Genética , Humanos , Camundongos , Camundongos SCID , Fenótipo , Retroviridae/genética , Células-Tronco/citologia , Fatores de Tempo , Transdução Genética
17.
Methods Enzymol ; 346: 573-89, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11883092

RESUMO

Oncoretroviral vectors and lentiviral vectors offer the potential for long-term gene expression by virtue of their stable chromosomal integration and lack of viral gene expression. Consequently, their integration allows passage of the transgene to all progeny cells, which makes them particularly suitable for stem cell transduction. However, a disadvantage of oncoretroviral vectors based on Moloney murine leukemia virus (MoMLV) is that cell division is required for transduction and integration, thereby limiting oncoretroviral-mediated gene therapy to actively dividing target cells. In contrast, lentiviral vectors can transduce both dividing and nondividing cells. Lentiviral vectors have been derived from either human or primate lentiviruses, with the human immunodeficiency virus (HIV) as prototype, or from nonprimate lentiviruses, such as the equine infectious anemia virus (EIAV). The ability to pseudotype oncoretroviral and lentiviral vectors with the vesicular stomatitis virus G glycoprotein (VSV-G) allowed for the production of high-titer vectors (10(9)-10(10) transducing units/ml). These high-titer vector preparations were shown to effectively cure genetic diseases in experimental animal models and constitute an essential step toward direct in vivo gene therapy applications. This chapter focuses on different methods that permit large-scale production of high-titer VSV-G pseudotyped oncoretroviral and primate or nonprimate lentiviral vectors and highlights their importance for achieving therapeutic effects in preclinical animal models.


Assuntos
Terapia Genética , Retroviridae/genética , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Humanos , Camundongos
18.
Methods Mol Biol ; 859: 241-54, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22367876

RESUMO

The PiggyBac (PB) transposon system was originally derived from the cabbage looper moth Trichoplusia ni and represents one of the most promising transposon systems to date. Engineering of the PB transposase enzyme (PBase) and its cognate transposon DNA elements resulted in a substantial increase in transposition activities. Consequently, this has greatly enhanced the versatility of the PB toolbox. It is now widely used for stable gene delivery into a broad variety of cell types from different species, including mammalian cells. This opened up new perspectives for potential therapeutic applications in the fields of gene therapy and regenerative medicine. In particular, we have recently demonstrated that PB transposons could be used to stably deliver genes into human CD34(+) hematopoietic stem cells (HSCs) resulting in sustained transgene expression in its differentiated progeny. The PB transposon system is particularly attractive for the generation of induced pluripotent stem cells (iPS). Typically, this can be accomplished by stable gene transfer of genes encoding one or more reprogramming factors (i.e., c-MYC, KLF-4, OCT-4, and/or SOX-2). We have generated a PB-based nonviral reprogramming toolbox that contains different combinations of these reprogramming genes. The main advantage of using this PB toolbox for iPS generation is that the reprogramming cassette can be excised by de novo transposase expression, without leaving any molecular trace in the target cell genome. This "traceless excision" paradigm obviates potential risks associated with inadvertent re-expression of reprogramming factors in the iPS progeny. These various applications in gene therapy, stem cell engineering, and regenerative medicine underscore the emerging versatility of the PB toolbox.


Assuntos
Terapia Genética/métodos , Animais , Técnicas de Cultura de Células , Elementos de DNA Transponíveis , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/transplante , Mutagênese Insercional , Transfecção , Transgenes
19.
Curr Gene Ther ; 11(5): 406-13, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21864290

RESUMO

Transposon technology is a particularly attractive non-viral gene delivery paradigm that allows for efficient genomic integration into a variety of different cell types. In particular, transposon-mediated gene transfer is a promising tool for stem cell research, by virtue of its ability to efficiently and stably transfer genes into adult and induced pluripotent stem (iPS) cells. Moreover, transposons open up new perspectives for non-viral-mediated stem cell-based gene therapy. Several transposon systems, especially the Sleeping Beauty (SB), the piggyBac (PB) and Tol2, have been optimized for gene transfer into mammalian cells. In particular, SB resulted in stable gene transfer into various adult stem cells including human CD34(+) hematopoietic stem cells (HSCs), myoblasts and mesenchymal stem cells (MSCs). This has been confirmed with PB, yielding stable gene transfer in human CD34(+) HSCs. Recently, PB transposons were used to deliver the genes encoding the reprogramming factors into somatic cells making it an attractive technology for generating iPS cells. Subsequent de novo expression of the PB transposase resulted in traceless excision of the reprogramming cassette. This prevented inadvertent re-expression of the reprogramming factors obviating some of the concerns associated with the use of integrating vectors. Transposons have also been used as a novel non-viral paradigm to coax differentiation of iPS cells into their desired target cells by forced expression of specific differentiation factors. This review focuses on the emerging potential of transposons for gene transfer into stem cells and its implications for gene therapy and regenerative medicine.


Assuntos
Elementos de DNA Transponíveis/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Vetores Genéticos , Células-Tronco Hematopoéticas/citologia , Humanos , Integrases/genética , Células-Tronco Mesenquimais/citologia , Transgenes , Transposases/genética
20.
Nat Genet ; 41(6): 753-61, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19412179

RESUMO

The Sleeping Beauty (SB) transposon is a promising technology platform for gene transfer in vertebrates; however, its efficiency of gene insertion can be a bottleneck in primary cell types. A large-scale genetic screen in mammalian cells yielded a hyperactive transposase (SB100X) with approximately 100-fold enhancement in efficiency when compared to the first-generation transposase. SB100X supported 35-50% stable gene transfer in human CD34(+) cells enriched in hematopoietic stem or progenitor cells. Transplantation of gene-marked CD34(+) cells in immunodeficient mice resulted in long-term engraftment and hematopoietic reconstitution. In addition, SB100X supported sustained (>1 year) expression of physiological levels of factor IX upon transposition in the mouse liver in vivo. Finally, SB100X reproducibly resulted in 45% stable transgenesis frequencies by pronuclear microinjection into mouse zygotes. The newly developed transposase yields unprecedented stable gene transfer efficiencies following nonviral gene delivery that compare favorably to stable transduction efficiencies with integrating viral vectors and is expected to facilitate widespread applications in functional genomics and gene therapy.


Assuntos
Evolução Molecular , Transposases/genética , Vertebrados/genética , Sequência de Aminoácidos , Animais , Sequência Conservada , Elementos de DNA Transponíveis/genética , Humanos , Camundongos , Camundongos Transgênicos/genética , Dados de Sequência Molecular , Filogenia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transposases/química
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa