Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 93(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31413132

RESUMO

Vaccines aimed at inducing T cell responses to protect against human immunodeficiency virus (HIV) infection have been under development for more than 15 years. Replication-defective adenovirus (rAd) vaccine vectors are at the forefront of this work and have been tested extensively in the simian immunodeficiency virus (SIV) challenge macaque model. Vaccination with rAd vectors coding for SIV Gag or other nonenvelope proteins induces T cell responses that control virus load but disappointingly is unsuccessful so far in preventing infection, and attention has turned to inducing antibodies to the envelope. However, here we report that Mauritian cynomolgus macaques (MCM), Macaca fascicularis, vaccinated with unmodified SIV gag alone in a DNA prime followed by an rAd boost exhibit increased protection from infection by repeated intrarectal challenge with low-dose SIVmac251. There was no evidence of infection followed by eradication. A significant correlation was observed between cytokine expression by CD4 T cells and delayed infection. Vaccination with gag fused to the ubiquitin gene or fragmented, designed to increase CD8 magnitude and breadth, did not confer resistance to challenge or enhance immunity. On infection, a significant reduction in peak virus load was observed in all vaccinated animals, including those vaccinated with modified gag These findings suggest that a nonpersistent viral vector vaccine coding for internal virus proteins may be able to protect against HIV type 1 (HIV-1) infection. The mechanisms are probably distinct from those of antibody-mediated virus neutralization or cytotoxic CD8 cell killing of virus-infected cells and may be mediated in part by CD4 T cells.IMPORTANCE The simian immunodeficiency virus (SIV) macaque model represents the best animal model for testing new human immunodeficiency virus type 1 (HIV-1) vaccines. Previous studies employing replication-defective adenovirus (rAd) vectors that transiently express SIV internal proteins induced T cell responses that controlled virus load but did not protect against virus challenge. However, we show for the first time that SIV gag delivered in a DNA prime followed by a boost with an rAd vector confers resistance to SIV intrarectal challenge. Other partially successful SIV/HIV-1 protective vaccines induce antibody to the envelope and neutralize the virus or mediate antibody-dependent cytotoxicity. Induction of CD8 T cells which do not prevent initial infection but eradicate infected cells before infection becomes established has also shown some success. In contrast, the vaccine described here mediates resistance by a different mechanism from that described above, which may reflect CD4 T cell activity. This could indicate an alternative approach for HIV-1 vaccine development.


Assuntos
Produtos do Gene gag/imunologia , Vacinas contra a SAIDS/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Vírus Defeituosos/genética , Vírus Defeituosos/imunologia , Produtos do Gene gag/genética , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Macaca fascicularis , Masculino , Vacinas contra a SAIDS/administração & dosagem , Vacinas contra a SAIDS/genética , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vacinação , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Carga Viral
2.
Mol Ther ; 27(10): 1706-1717, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31526597

RESUMO

The field of cell and gene therapy (GT) is expanding rapidly and there is undoubtedly a wave of enthusiasm and anticipation for what these treatments could achieve next. Here we assessed the worldwide landscape of GT assets currently in early clinical development (clinical trial phase 1/2 or about to enter clinical trial). We included all gene therapies, i.e., strategies that modify an individual's protein make-up by introducing exogenous nucleic acid or nucleic acid modifiers, regardless of delivery. Unmodified cell therapies, oncology therapies (reviewed elsewhere), and vaccine programs (distinct therapeutic strategy) were not included. Using a December 31, 2018 cutoff date, we identified 336 gene therapies being developed for 138 different indications covering 165 genetic targets. In all, we found that the early clinical GT landscape comprises a very disparate group of drug candidates in terms of indications, organizations, and delivery methods. We also highlight interesting trends, revealing the evolution of the field toward in vivo therapies and adeno-associated virus vector-based delivery systems. It will be interesting to witness what proportion of this current list effectively translates into new medicines.


Assuntos
Sistemas de Liberação de Medicamentos/classificação , Terapia Genética/métodos , Ensaios Clínicos como Assunto , Vetores Genéticos/administração & dosagem , Humanos , Terapia de Alvo Molecular
3.
Nucleic Acids Res ; 44(2): 744-60, 2016 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-26682797

RESUMO

Duchenne muscular dystrophy (DMD) is a genetic neuromuscular disorder caused by the absence of dystrophin. We developed a novel gene therapy approach based on the use of the piggyBac (PB) transposon system to deliver the coding DNA sequence (CDS) of either full-length human dystrophin (DYS: 11.1 kb) or truncated microdystrophins (MD1: 3.6 kb; MD2: 4 kb). PB transposons encoding microdystrophins were transfected in C2C12 myoblasts, yielding 65±2% MD1 and 66±2% MD2 expression in differentiated multinucleated myotubes. A hyperactive PB (hyPB) transposase was then deployed to enable transposition of the large-size PB transposon (17 kb) encoding the full-length DYS and green fluorescence protein (GFP). Stable GFP expression attaining 78±3% could be achieved in the C2C12 myoblasts that had undergone transposition. Western blot analysis demonstrated expression of the full-length human DYS protein in myotubes. Subsequently, dystrophic mesoangioblasts from a Golden Retriever muscular dystrophy dog were transfected with the large-size PB transposon resulting in 50±5% GFP-expressing cells after stable transposition. This was consistent with correction of the differentiated dystrophic mesoangioblasts following expression of full-length human DYS. These results pave the way toward a novel non-viral gene therapy approach for DMD using PB transposons underscoring their potential to deliver large therapeutic genes.


Assuntos
Elementos de DNA Transponíveis/genética , Distrofina/genética , Terapia Genética/métodos , Distrofia Muscular de Duchenne/patologia , Animais , Diferenciação Celular , Células Cultivadas , Cães , Distrofina/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Masculino , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/terapia , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Transfecção
4.
Mol Ther ; 23(1): 43-52, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25195597

RESUMO

Gene therapy is a promising emerging therapeutic modality for the treatment of cardiovascular diseases and hereditary diseases that afflict the heart. Hence, there is a need to develop robust cardiac-specific expression modules that allow for stable expression of the gene of interest in cardiomyocytes. We therefore explored a new approach based on a genome-wide bioinformatics strategy that revealed novel cardiac-specific cis-acting regulatory modules (CS-CRMs). These transcriptional modules contained evolutionary-conserved clusters of putative transcription factor binding sites that correspond to a "molecular signature" associated with robust gene expression in the heart. We then validated these CS-CRMs in vivo using an adeno-associated viral vector serotype 9 that drives a reporter gene from a quintessential cardiac-specific α-myosin heavy chain promoter. Most de novo designed CS-CRMs resulted in a >10-fold increase in cardiac gene expression. The most robust CRMs enhanced cardiac-specific transcription 70- to 100-fold. Expression was sustained and restricted to cardiomyocytes. We then combined the most potent CS-CRM4 with a synthetic heart and muscle-specific promoter (SPc5-12) and obtained a significant 20-fold increase in cardiac gene expression compared to the cytomegalovirus promoter. This study underscores the potential of rational vector design to improve the robustness of cardiac gene therapy.


Assuntos
Dependovirus/genética , Terapia Genética/métodos , Genoma , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miosinas Ventriculares/genética , Animais , Sítios de Ligação , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/terapia , Biologia Computacional , Citomegalovirus/química , Citomegalovirus/genética , Expressão Gênica , Engenharia Genética/métodos , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Miocárdio/patologia , Miócitos Cardíacos/patologia , Motivos de Nucleotídeos , Regiões Promotoras Genéticas , Ligação Proteica , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Miosinas Ventriculares/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(8): 3041-6, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23386724

RESUMO

Stabilization of virus protein structure and nucleic acid integrity is challenging yet essential to preserve the transcriptional competence of live recombinant viral vaccine vectors in the absence of a cold chain. When coupled with needle-free skin delivery, such a platform would address an unmet need in global vaccine coverage against HIV and other global pathogens. Herein, we show that a simple dissolvable microneedle array (MA) delivery system preserves the immunogenicity of vaccines encoded by live recombinant human adenovirus type 5 (rAdHu5). Specifically, dried rAdHu5 MA immunization induced CD8(+) T-cell expansion and multifunctional cytokine responses equipotent with conventional injectable routes of immunization. Intravital imaging demonstrated MA cargo distributed both in the epidermis and dermis, with acquisition by CD11c(+) dendritic cells (DCs) in the dermis. The MA immunizing properties were attributable to CD11c(+) MHCII(hi) CD8α(neg) epithelial cell adhesion molecule (EpCAM(neg)) CD11b(+) langerin (Lang; CD207)(neg) DCs, but neither Langerhans cells nor Lang(+) DCs were required for CD8(+) T-cell priming. This study demonstrates an important technical advance for viral vaccine vectors progressing to the clinic and provides insights into the mechanism of CD8(+) T-cell priming by live rAdHu5 MAs.


Assuntos
Adenoviridae/imunologia , Antígenos CD/fisiologia , Linfócitos T CD8-Positivos/imunologia , Lectinas Tipo C/fisiologia , Lectinas de Ligação a Manose/fisiologia , Agulhas , Pele , Vacinas Virais/imunologia , Adenoviridae/genética , Citometria de Fluxo , Vetores Genéticos , Microscopia Confocal
6.
Mol Ther ; 19(7): 1331-41, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21266958

RESUMO

There is great interest in transdifferentiating cells from one lineage into those of another and in dedifferentiating mature cells back into a stem/progenitor cell state by deploying naturally occurring transcription factors (TFs). Often, however, steering cellular differentiation pathways in a predictable and efficient manner remains challenging. Here, we investigated the principle of combining domains from different lineage-specific TFs to improve directed cellular differentiation. As proof-of-concept, we engineered the whole-human TF MyoDCD, which has the NH(2)-terminal transcription activation domain (TAD) and adjacent DNA-binding motif of MyoD COOH-terminally fused to the TAD of myocardin (MyoCD). We found via reporter gene and marker protein assays as well as by a cell fusion readout system that, targeting the TAD of MyoCD to genes normally responsive to the skeletal muscle-specific TF MyoD enforces more robust myogenic reprogramming of nonmuscle cells than that achieved by the parental, prototypic master TF, MyoD. Human mesenchymal stem cells (hMSCs) transduced with a codon-optimized microdystrophin gene linked to a synthetic striated muscle-specific promoter and/or with MyoD or MyoDCD were evaluated for complementing the genetic defect in Duchenne muscular dystrophy (DMD) myocytes through heterotypic cell fusion. Cotransduction of hMSCs with MyoDCD and microdystrophin led to chimeric myotubes containing the highest dystrophin levels.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Fatores de Transcrição/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Citometria de Fluxo , Humanos , Microscopia de Fluorescência , Fatores de Transcrição/genética
7.
Proc Natl Acad Sci U S A ; 106(47): 19940-5, 2009 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-19918060

RESUMO

In the recently halted HIV type 1 (HIV-1) vaccine STEP trial, individuals that were seropositive for adenovirus serotype 5 (Ad5) showed increased rates of HIV-1 infection on vaccination with an Ad5 vaccine. We propose that this was due to activation and expansion of Ad5-specific mucosal-homing memory CD4 T cells. To test this hypothesis, Ad5 and Ad11 antibody titers were measured in 20 healthy volunteers. Dendritic cells (DCs) from these individuals were pulsed with replication defective Ad5 or Ad11 and co-cultured with autologous lymphocytes. Cytokine profiles, proliferative capacity, mucosal migration potential, and susceptibility to HIV infection of the adenovirus-stimulated memory CD4 T cells were measured. Stimulation of T cells from healthy Ad5-seropositive but Ad11-seronegative individuals with Ad5, or serologically distinct Ad11 vectors induced preferential expansion of adenovirus memory CD4 T cells expressing alpha(4)beta(7) integrins and CCR9, indicating a mucosal-homing phenotype. CD4 T-cell proliferation and IFN-gamma production in response to Ad stimulation correlated with Ad5 antibody titers. However, Ad5 serostatus did not correlate with total cytokine production upon challenge with Ad5 or Ad11. Expanded Ad5 and Ad11 memory CD4 T cells showed an increase in CCR5 expression and higher susceptibility to infection by R5 tropic HIV-1. This suggests that adenoviral-based vaccination against HIV-1 in individuals with preexisting immunity against Ad5 results in preferential expansion of HIV-susceptible activated CD4 T cells that home to mucosal tissues, increases the number of virus targets, and leads to a higher susceptibility to HIV acquisition.


Assuntos
Adenoviridae/imunologia , Linfócitos T CD4-Positivos/imunologia , HIV-1/imunologia , Imunidade nas Mucosas/imunologia , Memória Imunológica/imunologia , Vacinação , Vacinas contra a AIDS/imunologia , Adenoviridae/genética , Anticorpos Antivirais/imunologia , Linfócitos T CD4-Positivos/citologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Infecções por HIV/imunologia , HIV-1/patogenicidade , Humanos , Integrina alfa4/imunologia , Cadeias beta de Integrinas/imunologia , Ativação Linfocitária/imunologia , Mucosa/imunologia , Fenótipo , Receptores CCR/imunologia , Receptores CCR4/imunologia
8.
Int J Cancer ; 129(7): 1541-51, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21455986

RESUMO

ß-catenin is a multifunctional protein identified to be pivotal in embryonic patterning, organogenesis and adult homeostasis. It plays a critical structural role in mediating cadherin junctions and is also an essential transcriptional co-activator in the canonical Wnt pathway. Evidence has been documented that both the canonical Wnt pathway and cadherin junctions are deregulated or impaired in a plethora of human malignancies. In the light of this, there has been a recent surge in elucidating the mechanisms underlying the etiology of cancer development from the perspective of ß-catenin. Here, we focus on the emerging roles of ß-catenin in the process of tumorigenesis by discussing novel functions of old players and new proteins, mechanisms identified to mediate or interact with ß-catenin and the most recently unraveled clinical implications of ß-catenin regulatory pathways toward tumor suppression.


Assuntos
Neoplasias/etiologia , beta Catenina/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína da Polipose Adenomatosa do Colo/metabolismo , Proteína Axina , Adesão Celular , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Proteínas Relacionadas a Receptor de LDL/metabolismo , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Terapia de Alvo Molecular , Metástase Neoplásica , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/antagonistas & inibidores
9.
J Gene Med ; 13(9): 497-506, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22144143

RESUMO

BACKGROUND: Duchenne muscular dystrophy (DMD) is a severe, inherited, muscle-wasting disorder caused by mutations in the dystrophin gene. Preclinical studies of adeno-associated virus gene therapy for DMD have been described in mouse and dog models of this disease. However, low and transient expression of microdystrophin in dystrophic dogs and a lack of long-term microdystrophin expression associated with a CD8(+) T-cell response in DMD patients suggests that the development of improved microdystrophin genes and delivery strategies is essential for successful clinical trials in DMD patients. METHODS: We have previously shown the efficiency of mRNA sequence optimization of mouse microdystrophin in ameliorating the pathology of dystrophic mdx mice. In the present study, we generated adeno-associated virus (AAV)2/8 vectors expressing an mRNA sequence-optimized canine microdystrophin under the control of a muscle-specific promoter and injected intramuscularly into a single canine X-linked muscular dystrophy (CXMDj) dog. RESULTS: Expression of stable and high levels of microdystrophin was observed along with an association of the dystrophin-associated protein complex in intramuscularly injected muscles of a CXMDj dog for at least 8 weeks without immune responses. Treated muscles were highly protected from dystrophic damage, with reduced levels of myofiber permeability and central nucleation. CONCLUSIONS: The data obtained in the present study suggest that the use of canine-specific and mRNA sequence-optimized microdystrophin genes in conjunction with a muscle-specific promoter results in high and stable levels of microdystrophin expression in a canine model of DMD. This approach will potentially allow the reduction of dosage and contribute towards the development of a safe and effective AAV gene therapy clinical trial protocol for DMD.


Assuntos
Dependovirus/genética , Distrofina/metabolismo , Terapia Genética/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , RNA Mensageiro/metabolismo , Animais , Primers do DNA/genética , Cães , Distrofina/genética , Vetores Genéticos/administração & dosagem , Técnicas Histológicas , Injeções Intramusculares , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/metabolismo
10.
RSC Med Chem ; 12(3): 330-352, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34046619

RESUMO

Cell and gene therapies have achieved impressive results in the treatment of rare genetic diseases using gene corrected stem cells and haematological cancers using chimeric antigen receptor T cells. However, these two fields face significant challenges such as demonstrating long-term efficacy and safety, and achieving cost-effective, scalable manufacturing processes. The use of small molecules is a key approach to overcome these barriers and can benefit cell and gene therapies at multiple stages of their lifecycle. For example, small molecules can be used to optimise viral vector production during manufacturing or used in the clinic to enhance the resistance of T cell therapies to the immunosuppressive tumour microenvironment. Here, we review current uses of small molecules in cell and gene therapy and highlight opportunities for medicinal chemists to further consolidate the success of cell and gene therapies.

11.
Hum Gene Ther ; 32(19-20): 1120-1137, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34662232

RESUMO

Gene therapy is a relatively novel field that amounts to around four decades of continuous growth with its good and bad moments. Currently, the field has entered the clinical arena with the ambition to fulfil its promises for a permanent fix of incurable genetic disorders. Hemoglobinopathies as target diseases and hematopoietic stem cells (HSCs) as target cells of genetic interventions had a major share in the research effort toward efficiently implementing gene therapy. Dissection of HSC biology and improvements in gene transfer and gene expression technologies evolved in an almost synchronous manner to a point where the two fields seem to be functionally intercalated. In this review, we focus specifically on the development of gene therapy for hemoglobin disorders and look at both gene addition and gene correction strategies that may dominate the field of HSC-directed gene therapy in the near future and transform the therapeutic landscape for genetic diseases.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Hemoglobinopatias , Edição de Genes , Terapia Genética , Vetores Genéticos , Células-Tronco Hematopoéticas , Hemoglobinopatias/genética , Hemoglobinopatias/terapia , Humanos
12.
BMC Genomics ; 11: 345, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20515474

RESUMO

BACKGROUND: Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disorder caused by mutations in the dystrophin gene. DMD has a complex and as yet incompletely defined molecular pathophysiology hindering development of effective ameliorative approaches. Transcriptomic studies so far conducted on dystrophic cells and tissues suffer from non-specific changes and background noise due to heterogeneous comparisons and secondary pathologies. A study design in which a perfectly matched control cell population is used as reference for transcriptomic studies will give a much more specific insight into the effects of dystrophin deficiency and DMD pathophysiology. RESULTS: Using RNA interference (RNAi) to knock down dystrophin in myotubes from C57BL10 mice, we created a homogenous model to study the transcriptome of dystrophin-deficient myotubes. We noted significant differences in the global gene expression pattern between these myotubes and their matched control cultures. In particular, categorical analyses of the dysregulated genes demonstrated significant enrichment of molecules associated with the components of muscle cell contractile unit, ion channels, metabolic pathways and kinases. Additionally, some of the dysregulated genes could potentially explain conditions and endophenotypes associated with dystrophin deficiency, such as dysregulation of calcium homeostasis (Pvalb and Casq1), or cardiomyopathy (Obscurin, Tcap). In addition to be validated by qPCR, our data gains another level of validity by affirmatively reproducing several independent studies conducted previously at genes and/or protein levels in vivo and in vitro. CONCLUSION: Our results suggest that in striated muscles, dystrophin is involved in orchestrating proper development and organization of myofibers as contractile units, depicting a novel pathophysiology for DMD where the absence of dystrophin results in maldeveloped myofibers prone to physical stress and damage. Therefore, it becomes apparent that any gene therapy approaches for DMD should target early stages in muscle development to attain a maximum clinical benefit. With a clear and specific definition of the transcriptome of dystrophin deficiency, manipulation of identified dysregulated molecules downstream of dystrophin may lead to novel ameliorative approaches for DMD.


Assuntos
Diferenciação Celular/genética , Distrofina/deficiência , Distrofina/genética , Perfilação da Expressão Gênica , Contração Muscular/genética , Músculos/metabolismo , Interferência de RNA , Animais , Células Cultivadas , Técnicas de Silenciamento de Genes , Loci Gênicos/genética , Camundongos , MicroRNAs/genética , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/fisiologia , Músculos/citologia , Músculos/fisiologia , Miocárdio/metabolismo , Fenótipo
13.
Hum Mol Genet ; 17(17): 2622-32, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18511456

RESUMO

Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disorder caused by mutations in the dystrophin gene. DMD has a complex and as yet incompletely defined molecular pathophysiology. The peak of the pathology attributed to dystrophin deficiency happens between 3 and 8 weeks of age in mdx mice, the animal model of DMD. Accordingly, we hypothesized that the pathology observed with dystrophin deficiency may be developmentally regulated. Initially, we demonstrated that profound small interfering RNA-mediated dystrophin knockdown could be achieved in mouse primary muscle cultures. The use of adeno-associated virus vectors to express short-hairpin RNAs targeting dystrophin in skeletal muscle in vivo yielded a potent and specific dystrophin knockdown, but only after approximately 5 months, indicating the very long half-life of dystrophin. Interestingly, and in contrast to what is observed in congenital dystrophin deficiency, long-term ( approximately 1 year) dystrophin knockdown in adult mice did not result, per se, in overt dystrophic pathology or upregulation of utrophin. This supports our hypothesis and suggests new pathophysiology of the disease. Furthermore, taking into account the rather long half-life of dystrophin, and the notion that the development of pathology is age-dependent, it indicates that a single gene therapy approach before the onset of pathology might convey a long-term cure for DMD.


Assuntos
Distrofina/genética , Distrofia Muscular Animal/fisiopatologia , Interferência de RNA , Animais , Células Cultivadas , Distrofina/metabolismo , Metenamina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA Interferente Pequeno/metabolismo
14.
Mol Ther ; 16(11): 1825-32, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18766174

RESUMO

Duchenne muscular dystrophy is a fatal muscle-wasting disorder. Lack of dystrophin compromises the integrity of the sarcolemma and results in myofibers that are highly prone to contraction-induced injury. Recombinant adeno-associated virus (rAAV)-mediated dystrophin gene transfer strategies to muscle for the treatment of Duchenne muscular dystrophy (DMD) have been limited by the small cloning capacity of rAAV vectors and high titers necessary to achieve efficient systemic gene transfer. In this study, we assess the impact of codon optimization on microdystrophin (DeltaAB/R3-R18/DeltaCT) expression and function in the mdx mouse and compare the function of two different configurations of codon-optimized microdystrophin genes (DeltaAB/R3-R18/DeltaCT and DeltaR4-R23/DeltaCT) under the control of a muscle-restrictive promoter (Spc5-12). Codon optimization of microdystrophin significantly increases levels of microdystrophin mRNA and protein after intramuscular and systemic administration of plasmid DNA or rAAV2/8. Physiological assessment demonstrates that codon optimization of DeltaAB/R3-R18/DeltaCT results in significant improvement in specific force, but does not improve resistance to eccentric contractions compared with noncodon-optimized DeltaAB/R3-R18/DeltaCT. However, codon-optimized microdystrophin DeltaR4-R23/DeltaCT completely restored specific force generation and provided substantial protection from contraction-induced injury. These results demonstrate that codon optimization of microdystrophin under the control of a muscle-specific promoter can significantly improve expression levels such that reduced titers of rAAV vectors will be required for efficient systemic administration.


Assuntos
Códon , Dependovirus/genética , Distrofina/genética , Distrofia Muscular Animal/terapia , RNA Mensageiro/genética , Animais , Distrofina/biossíntese , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/fisiopatologia , Transgenes
15.
Immunol Lett ; 115(2): 117-25, 2008 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-18079003

RESUMO

Vaccine design approaches that target dendritic cells (DC) aim at achieving high levels of transgene expression. Careful selection of the promoter element driving the foreign gene is therefore important. We have constructed adenovirus vectors carrying the gene for enhanced green fluorescent protein (eGFP) driven by three different promoters, CMV, CMV5 and ubiquitin C (UbC) promoter, and analysed their activity in different populations of human DC, namely blood plasmacytoid (pDC) and myeloid DC (mDC), monocyte-derived DC (moDC), Langerhans (LC) and dermal type DC (dDC). Although the CMV5 promoter was more active than the other two promoters in the HeLa and 911HER cell lines, in human DC the highest level of transgene expression was seen with the CMV promoter. There was very low-level eGFP expression in all cell types transduced with the UbC promoter. Highest eGFP expression levels were observed in moDC, cultured mDC and LC and the lowest levels in pDC. Expression of eGFP was augmented in all DC populations upon stimulation with CD40 ligand (CD40L). These findings demonstrate that the CMV promoter is the most effective of the three promoters tested in a range of different human DC populations.


Assuntos
Células Dendríticas/metabolismo , Vetores Genéticos , Regiões Promotoras Genéticas , Transgenes , Vacinas , Adenoviridae/genética , Linhagem Celular Tumoral , Citomegalovirus/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Células de Langerhans/metabolismo , Transdução Genética , Transfecção , Ubiquitina C/genética
16.
Hematol Oncol Clin North Am ; 31(5): 753-770, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28895845

RESUMO

Gene delivery vectors that do not rely on host cell genome integration offer several advantages for gene transfer, chiefly the avoidance of insertional mutagenesis and position effect variegation. However, unless engineered for replication and segregation, nonintegrating vectors will dilute progressively in proliferating cells, and are not exempt of epigenetic effects. This article provides an overview of the main nonintegrating viral (adenoviral, adeno-associated viral, integration-deficient retro-lentiviral, poxviral), and nonviral (plasmid vectors, artificial chromosomes) vectors used for preclinical and clinical cell and gene therapy applications. Particular emphasis is placed on their use in hematologic disease.


Assuntos
Terapia Genética , Vetores Genéticos/genética , Adenoviridae/genética , Animais , Ensaios Clínicos como Assunto/história , Dependovirus/genética , Edição de Genes , Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética/efeitos adversos , Terapia Genética/história , Terapia Genética/métodos , Vetores Genéticos/classificação , História do Século XX , História do Século XXI , Humanos , Plasmídeos/genética , Poxviridae/genética , Transdução Genética
17.
Nat Commun ; 8: 16105, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28742067

RESUMO

Duchenne muscular dystrophy (DMD) is an incurable X-linked muscle-wasting disease caused by mutations in the dystrophin gene. Gene therapy using highly functional microdystrophin genes and recombinant adeno-associated virus (rAAV) vectors is an attractive strategy to treat DMD. Here we show that locoregional and systemic delivery of a rAAV2/8 vector expressing a canine microdystrophin (cMD1) is effective in restoring dystrophin expression and stabilizing clinical symptoms in studies performed on a total of 12 treated golden retriever muscular dystrophy (GRMD) dogs. Locoregional delivery induces high levels of microdystrophin expression in limb musculature and significant amelioration of histological and functional parameters. Systemic intravenous administration without immunosuppression results in significant and sustained levels of microdystrophin in skeletal muscles and reduces dystrophic symptoms for over 2 years. No toxicity or adverse immune consequences of vector administration are observed. These studies indicate safety and efficacy of systemic rAAV-cMD1 delivery in a large animal model of DMD, and pave the way towards clinical trials of rAAV-microdystrophin gene therapy in DMD patients.


Assuntos
Distrofina/genética , Técnicas de Transferência de Genes , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/genética , Administração Intravenosa , Animais , Dependovirus , Modelos Animais de Doenças , Cães , Terapia Genética , Vetores Genéticos , Masculino , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/fisiopatologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatologia , Transgenes
18.
Curr Gene Ther ; 15(4): 395-415, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26159373

RESUMO

Duchenne muscular dystrophy (DMD), an X-linked inherited musclewasting disease primarily affecting young boys with prevalence of between1:3,500- 1:5,000, is a rare genetic disease caused by defects in the gene for dystrophin. Dystrophin protein is critical to the stability of myofibers in skeletal and cardiac muscle. There is currently no cure available to ameliorate DMD and/or its patho-physiology. A number of therapeutic strategies including molecular-based therapeutics that replace or correct the missing or nonfunctional dystrophin protein have been devised to correct the patho-physiological consequences induced by dystrophin absence. We will review the current in vivo experimentation status (including preclinical models and clinical trials) for two of these approaches, namely: 1) Adeno-associated virus (AAV) mediated (micro) dystrophin gene augmentation/ supplementation and 2) Antisense oligonucleotide (AON)-mediated exon skipping strategies.


Assuntos
Dependovirus/genética , Distrofina/genética , Terapia Genética/métodos , Distrofia Muscular de Duchenne/etiologia , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos Antissenso/farmacologia , Animais , Ensaios Clínicos como Assunto , Dependovirus/imunologia , Modelos Animais de Doenças , Distrofina/deficiência , Éxons , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Humanos , Distrofia Muscular de Duchenne/genética , Oligonucleotídeos Antissenso/genética , Oxidiazóis/farmacologia , Edição de RNA
19.
Nat Commun ; 6: 7205, 2015 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-26018658

RESUMO

Myotonic Dystrophy type 1 (DM1) is a dominant neuromuscular disease caused by nuclear-retained RNAs containing expanded CUG repeats. These toxic RNAs alter the activities of RNA splicing factors resulting in alternative splicing misregulation and muscular dysfunction. Here we show that the abnormal splicing of DMD exon 78 found in dystrophic muscles of DM1 patients is due to the functional loss of MBNL1 and leads to the re-expression of an embryonic dystrophin in place of the adult isoform. Forced expression of embryonic dystrophin in zebrafish using an exon-skipping approach severely impairs the mobility and muscle architecture. Moreover, reproducing Dmd exon 78 missplicing switch in mice induces muscle fibre remodelling and ultrastructural abnormalities including ringed fibres, sarcoplasmic masses or Z-band disorganization, which are characteristic features of dystrophic DM1 skeletal muscles. Thus, we propose that splicing misregulation of DMD exon 78 compromises muscle fibre maintenance and contributes to the progressive dystrophic process in DM1.


Assuntos
Distrofina/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/genética , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/genética , Distrofia Miotônica/genética , Splicing de RNA/genética , Proteínas de Ligação a RNA/genética , Proteínas de Peixe-Zebra/genética , Animais , Cromatografia Líquida , Distrofina/metabolismo , Éxons , Homeostase , Humanos , Imuno-Histoquímica , Imunoprecipitação , Proteínas de Membrana/metabolismo , Camundongos , Microscopia Eletrônica , Fibras Musculares Esqueléticas/ultraestrutura , Proteínas Musculares/metabolismo , Distrofia Miotônica/patologia , Reação em Cadeia da Polimerase em Tempo Real , Retículo Sarcoplasmático/ultraestrutura , Espectrometria de Massas em Tandem , Proteínas de Peixe-Zebra/metabolismo
20.
Hum Gene Ther ; 25(2): 98-108, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24191945

RESUMO

Recombinant adeno-associated virus (rAAV) vectors have been shown to permit very efficient widespread transgene expression in skeletal muscle after systemic delivery, making these increasingly attractive as vectors for Duchenne muscular dystrophy (DMD) gene therapy. DMD is a severe muscle-wasting disorder caused by DMD gene mutations leading to complete loss of dystrophin protein. One of the major issues associated with delivery of the DMD gene, as a therapeutic approach for DMD, is its large open reading frame (ORF; 11.1 kb). A series of truncated microdystrophin cDNAs (delivered via a single AAV) and minidystrophin cDNAs (delivered via dual-AAV trans-spliced/overlapping reconstitution) have thus been extensively tested in DMD animal models. However, critical rod and hinge domains of dystrophin required for interaction with components of the dystrophin-associated protein complex, such as neuronal nitric oxide synthase, syntrophin, and dystrobrevin, are missing; these dystrophin domains may still need to be incorporated to increase dystrophin functionality and stabilize membrane rigidity. Full-length DMD gene delivery using AAV vectors remains elusive because of the limited single-AAV packaging capacity (4.7 kb). Here we developed a novel method for the delivery of the full-length DMD coding sequence to skeletal muscles in dystrophic mdx mice using a triple-AAV trans-splicing vector system. We report for the first time that three independent AAV vectors carrying "in tandem" sequential exonic parts of the human DMD coding sequence enable the expression of the full-length protein as a result of trans-splicing events cojoining three vectors via their inverted terminal repeat sequences. This method of triple-AAV-mediated trans-splicing could be applicable to the delivery of any large therapeutic gene (≥11 kb ORF) into postmitotic tissues (muscles or neurons) for the treatment of various inherited metabolic and genetic diseases.


Assuntos
Dependovirus/genética , Distrofina/genética , Vetores Genéticos/genética , Distrofia Muscular Animal/genética , Trans-Splicing , Animais , Sequência de Bases , Dependovirus/química , Amplificação de Genes , Expressão Gênica , Ordem dos Genes , Técnicas de Transferência de Genes , Vetores Genéticos/química , Humanos , Masculino , Camundongos , Camundongos Endogâmicos mdx , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Fases de Leitura Aberta , Alinhamento de Sequência , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA