Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Chem Phys ; 158(20)2023 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-37218701

RESUMO

We explore the relationship between a machine-learned structural quantity (softness) and excess entropy in simulations of supercooled liquids. Excess entropy is known to scale well the dynamical properties of liquids, but this quasi-universal scaling is known to breakdown in supercooled and glassy regimes. Using numerical simulations, we test whether a local form of the excess entropy can lead to predictions similar to those made by softness, such as the strong correlation with particles' tendency to rearrange. In addition, we explore leveraging softness to compute excess entropy in the traditional fashion over softness groupings. Our results show that the excess entropy computed over softness-binned groupings is correlated with activation barriers to rearrangement.

2.
Lab Anim (NY) ; 44(1): 32-8, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25526057

RESUMO

Within the biomedical research industry, people who work with laboratory animals may be at risk of developing laboratory animal allergy, which can lead to occupational asthma. Under UK and EU laws, employers must prevent or adequately control exposure to any hazardous substance, which includes animal allergens, so far as reasonably practicable, for the protection of all people on the premises. This can be achieved in part by reviewing the risk of allergen exposure in specific areas of a facility and implementing appropriate infrastructure, environmental and performance controls to minimize that risk. The authors describe the approach used at their institution to stratify risk of allergen exposure in various areas of the animal facility and to implement appropriate controls. They also discuss their use of a monitoring program to evaluate allergen concentrations in low- and high-risk areas of the animal facility and explain how the monitoring results can be applied to determine which controls are needed to minimize risk of exposure and to provide a safe working environment.


Assuntos
Alérgenos , Exposição Ocupacional/prevenção & controle , Animais , Animais de Laboratório , Fiscalização e Controle de Instalações , Humanos , Saúde Ocupacional , Medição de Risco
3.
Methods Mol Biol ; 709: 153-78, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21194027

RESUMO

Duchenne muscular dystrophy (DMD) is caused by mutations that disrupt the reading frame of the human DMD gene. Selective removal of exons flanking an out-of-frame DMD mutation can result in an in-frame mRNA transcript that may be translated into an internally deleted, Becker muscular dystrophy (BMD)-like, but functionally active dystrophin protein with therapeutic activity. Antisense oligonucleotides (AOs) can be designed to bind to complementary sequences in the targeted mRNA and modify pre-mRNA splicing to correct the reading frame of a mutated transcript so that gene expression is restored. AO-induced exon skipping producing functional truncated dystrophin exon has been demonstrated in animal models of DMD both in vitro and in vivo, and in DMD patient cells in vitro in culture, and in DMD muscle explants. More recently, AO-mediated exon skipping has been confirmed in DMD patients in Phase I clinical trials. However, it should be noted that personalized molecular medicine may be necessary, since the various reading frame-disrupting mutations are spread across the DMD gene. The different deletions that cause DMD would require skipping of different exons, which would require the optimization and clinical trial workup of many specific AOs. This chapter describes the methodologies available for the optimization of AOs, and in particular phosphorodiamidate morpholino oligomers (PMOs), for the targeted skipping of specific exons on the DMD gene.


Assuntos
DNA Antissenso/uso terapêutico , Distrofina/genética , Morfolinas/uso terapêutico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Splicing de RNA , Processamento Alternativo , Biologia Computacional , Distrofina/metabolismo , Éxons , Mutação da Fase de Leitura , Expressão Gênica , Terapia Genética/métodos , Humanos , Morfolinos , Músculo Esquelético/fisiologia , Fases de Leitura
4.
Metabolism ; 60(4): 491-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20580777

RESUMO

Intramuscular injection of adeno-associated viral (AAV) vectors is potentially a safe, minimally invasive procedure for the long-term gene expression of circulating antiatherogenic proteins. Here, we compare secretion and atheroprotective effects of human apoE3 after injection of 3 pseudotyped AAV vectors (AAV2/7, AAV2/8, or AAV2/9), driven by the CMV enhancer/chicken ß-actin (CAG) promoter, into skeletal muscle of hyperlipidemic apolipoprotein E-deficient (apoE⁻/⁻) mice. Vector viabilities were verified by transducing cultured C2C12 mouse myotubes and assessing secretion of human apoE3 protein. Both hind limb tibialis anterior muscles of female C57BL/6 apoE⁻/⁻ mice, 2 months old and fed a high-fat diet, were each injected with 1 x 10¹° vector genomes of AAV vector. Identical noninjected mice served as controls; and blood was collected at weeks 0, 1, 2, 4, and 13. At termination (13 weeks), the brachiocephalic artery was excised; and after staining sections, plaque morphometry and fractional lipid content were quantified by computerized image analysis. Intramuscular injection of AAV2/7 and AAV2/8 vectors produced up to 2 µg human apoE3 per milliliter plasma, just below the threshold to reverse dyslipoproteinemia. AAV2/9 was notably less effective, mice having a 3-fold lower level of plasma apoE3 at 13 weeks and a 50% greater burden of atherosclerotic plaque lipid in their brachiocephalic arteries. We conclude that although vector refinement is needed to exploit fully apoE3 atheroprotective functions, AAV2/7 and AAV2/8 are promising gene transfer vectors for muscle-based expression of antiatherogenic circulating proteins.


Assuntos
Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Aterosclerose/prevenção & controle , Dependovirus/genética , Músculo Esquelético/metabolismo , Animais , Aterosclerose/metabolismo , Linhagem Celular , Artérias Cerebrais/patologia , Meios de Cultura , Dependovirus/classificação , Dependovirus/imunologia , Ensaio de Imunoadsorção Enzimática , Terapia Genética , Vetores Genéticos , Humanos , Hipercolesterolemia/terapia , Metabolismo dos Lipídeos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sorotipagem , Transdução Genética
5.
Mol Ther ; 19(2): 345-54, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21102560

RESUMO

The administration of antisense oligonucleotides (AOs) to skip one or more exons in mutated forms of the DMD gene and so restore the reading frame of the transcript is one of the most promising approaches to treat Duchenne muscular dystrophy (DMD). At present, preclinical studies demonstrating the efficacy and safety of long-term AO administration have not been conducted. Furthermore, it is essential to determine the minimal effective dose and frequency of administration. In this study, two different low doses (LDs) of phosphorodiamidate morpholino oligomer (PMO) designed to skip the mutated exon 23 in the mdx dystrophic mouse were administered for up to 12 months. Mice treated for 50 weeks showed a substantial dose-related amelioration of the pathology, particularly in the diaphragm. Moreover, the generalized physical activity was profoundly enhanced compared to untreated mdx mice showing that widespread, albeit partial, dystrophin expression restores the normal activity in mdx mice. Our results show for the first time that a chronic long-term administration of LDs of unmodified PMO, equivalent to doses in use in DMD boys, is safe, significantly ameliorates the muscular dystrophic phenotype and improves the activity of dystrophin-deficient mice, thus encouraging the further clinical translation of this approach in humans.


Assuntos
Morfolinas/uso terapêutico , Distrofia Muscular de Duchenne/tratamento farmacológico , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Western Blotting , Creatina Quinase/sangue , Creatinina/sangue , Eletrofisiologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos mdx , Morfolinos , Distrofia Muscular de Duchenne/sangue , gama-Glutamiltransferase
6.
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
7.
Neuromuscul Disord ; 20(2): 102-10, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20079639

RESUMO

Duchenne muscular dystrophy (DMD) is caused by the lack of functional dystrophin protein, most commonly as a result of a range of out-of-frame mutations in the DMD gene. Modulation of pre-mRNA splicing with antisense oligonucleotides (AOs) to restore the reading frame has been demonstrated in vitro and in vivo, such that truncated but functional dystrophin is expressed. AO-induced skipping of exon 51 of the DMD gene, which could treat 13% of DMD patients, has now progressed to clinical trials. We describe here the methodical, cooperative comparison, in vitro (in DMD cells) and in vivo (in a transgenic mouse expressing human dystrophin), of 24 AOs of the phosphorodiamidate morpholino oligomer (PMO) chemistry designed to target exon 53 of the DMD gene, skipping of which could be potentially applicable to 8% of patients. A number of the PMOs tested should be considered worthy of development for clinical trial.


Assuntos
Distrofina/efeitos dos fármacos , Éxons/efeitos dos fármacos , Marcação de Genes/métodos , Terapia Genética/métodos , Distrofia Muscular de Duchenne/tratamento farmacológico , Oligonucleotídeos Antissenso/farmacologia , Animais , Sequência de Bases/efeitos dos fármacos , Sequência de Bases/genética , Células Cultivadas , Modelos Animais de Doenças , Distrofina/química , Distrofina/genética , Éxons/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Morfolinas/química , Morfolinas/farmacologia , Morfolinas/uso terapêutico , Morfolinos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Mutação/genética , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/uso terapêutico , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética
8.
In Vivo ; 23(6): 885-93, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20023229

RESUMO

UNLABELLED: To improve liver-directed retroviral-mediated gene transfer, we injected C57/BL10 mice intravenously with three adenoviral vectors encoding retroviral vector genome and structural components: AdGagPol expressing the respective structural genes of Moloney murine leukaemia virus, Ad10A1Env expressing the 10A1 envelope protein of 10A1-MuLV, and AdLEIN, encoding the LEIN retrovirus genome, expressing green fluorescence protein (eGFP) and the neomycin resistance gene. MATERIALS AND METHODS: The extent of eGFP expression was determined after 1 and 15 weeks by fluorescence microscopy and FACS analysis. Proviral integration was determined by a novel PCR-based technique. RESULTS: Hepatocytes infected with all three Ad vectors generated LEIN retrovirus after one week and in situ transduction of neighbouring cells resulted in stable proviral integration associated with eGFP expression ranging from 4.3% to 20.5% in different liver cell populations 15 weeks post-infection. CONCLUSION: Hybrid adeno-retroviral vectors can be efficiently used to improve the efficiency of retroviral-mediated gene transfer to the liver.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Hepatócitos/virologia , Vírus da Leucemia Murina de Moloney/genética , Animais , Linhagem Celular Tumoral , Terapia Genética , Proteínas de Fluorescência Verde/metabolismo , Hepatócitos/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Transdução Genética , Integração Viral
9.
Lancet Neurol ; 8(10): 918-28, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19713152

RESUMO

BACKGROUND: Mutations that disrupt the open reading frame and prevent full translation of DMD, the gene that encodes dystrophin, underlie the fatal X-linked disease Duchenne muscular dystrophy. Oligonucleotides targeted to splicing elements (splice switching oligonucleotides) in DMD pre-mRNA can lead to exon skipping, restoration of the open reading frame, and the production of functional dystrophin in vitro and in vivo, which could benefit patients with this disorder. METHODS: We did a single-blind, placebo-controlled, dose-escalation study in patients with DMD recruited nationally, to assess the safety and biochemical efficacy of an intramuscular morpholino splice-switching oligonucleotide (AVI-4658) that skips exon 51 in dystrophin mRNA. Seven patients with Duchenne muscular dystrophy with deletions in the open reading frame of DMD that are responsive to exon 51 skipping were selected on the basis of the preservation of their extensor digitorum brevis (EDB) muscle seen on MRI and the response of cultured fibroblasts from a skin biopsy to AVI-4658. AVI-4658 was injected into the EDB muscle; the contralateral muscle received saline. Muscles were biopsied between 3 and 4 weeks after injection. The primary endpoint was the safety of AVI-4658 and the secondary endpoint was its biochemical efficacy. This trial is registered, number NCT00159250. FINDINGS: Two patients received 0.09 mg AVI-4658 in 900 microL (0.9%) saline and five patients received 0.9 mg AVI-4658 in 900 microL saline. No adverse events related to AVI-4658 administration were reported. Intramuscular injection of the higher-dose of AVI-4658 resulted in increased dystrophin expression in all treated EDB muscles, although the results of the immunostaining of EDB-treated muscle for dystrophin were not uniform. In the areas of the immunostained sections that were adjacent to the needle track through which AVI-4658 was given, 44-79% of myofibres had increased expression of dystrophin. In randomly chosen sections of treated EDB muscles, the mean intensity of dystrophin staining ranged from 22% to 32% of the mean intensity of dystrophin in healthy control muscles (mean 26.4%), and the mean intensity was 17% (range 11-21%) greater than the intensity in the contralateral saline-treated muscle (one-sample paired t test p=0.002). In the dystrophin-positive fibres, the intensity of dystrophin staining was up to 42% of that in healthy muscle. We showed expression of dystrophin at the expected molecular weight in the AVI-4658-treated muscle by immunoblot. INTERPRETATION: Intramuscular AVI-4658 was safe and induced the expression of dystrophin locally within treated muscles. This proof-of-concept study has led to an ongoing systemic clinical trial of AVI-4658 in patients with DMD. FUNDING: UK Department of Health.


Assuntos
Distrofina/biossíntese , Terapia Genética/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos/administração & dosagem , Adolescente , Criança , Relação Dose-Resposta a Droga , Distrofina/genética , Humanos , Imuno-Histoquímica , Masculino , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Oligonucleotídeos/efeitos adversos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Método Simples-Cego
10.
Rejuvenation Res ; 12(2): 85-94, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19405813

RESUMO

Myostatin is a member of the transformating growth factor-beta (TGF-beta) superfamily of proteins and is produced almost exclusively in skeletal muscle tissue, where it is secreted and circulates as a serum protein. Myostatin acts as a negative regulator of muscle mass through the canonical SMAD2/3/4 signaling pathway. Naturally occurring myostatin mutants exhibit a 'double muscling' phenotype in which muscle mass is dramatically increased as a result of both hypertrophy and hyperplasia. Myostatin is naturally inhibited by its own propeptide; therefore, we assessed the impact of adeno-associated virus-8 (AAV8) myostatin propeptide vectors when systemically introduced in MF-1 mice. We noted a significant systemic increase in muscle mass in both slow and fast muscle phenotypes, with no evidence of hyperplasia; however, the nuclei-to- cytoplasm ratio in all myofiber types was significantly reduced. An increase in muscle mass in slow (soleus) muscle led to an increase in force output; however, an increase in fast (extensor digitorum longus [EDL]) muscle mass did not increase force output. These results suggest that the use of gene therapeutic regimens of myostatin inhibition for age-related or disease-related muscle loss may have muscle-specific effects.


Assuntos
Dependovirus/metabolismo , Técnicas de Transferência de Genes , Fibras Musculares de Contração Rápida/fisiologia , Fibras Musculares de Contração Lenta/fisiologia , Miostatina/administração & dosagem , Peptídeos/administração & dosagem , Precursores de Proteínas/administração & dosagem , Animais , Fenômenos Biomecânicos/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Hiperplasia , Hipertrofia , Injeções Intravenosas , Camundongos , Fibras Musculares de Contração Rápida/efeitos dos fármacos , Fibras Musculares de Contração Rápida/patologia , Fibras Musculares de Contração Lenta/efeitos dos fármacos , Fibras Musculares de Contração Lenta/patologia , Miostatina/antagonistas & inibidores , Miostatina/metabolismo , Miostatina/farmacologia , Tamanho do Órgão/efeitos dos fármacos , Peptídeos/metabolismo , Peptídeos/farmacologia , Precursores de Proteínas/metabolismo , Precursores de Proteínas/farmacologia , Células Satélites de Músculo Esquelético/efeitos dos fármacos , Células Satélites de Músculo Esquelético/patologia
11.
Hum Gene Ther ; 20(9): 955-65, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19469709

RESUMO

Duchenne muscular dystrophy (DMD) is a myodegenerative disorder caused primarily by mutations that create premature termination of dystrophin translation. The antisense oligonucleotide approach for skipping dystrophin exons allows restoration of the correct reading frame in the dystrophin transcript, thus producing a shorter protein. A similar approach in humans would result in the conversion of DMD to the milder Becker muscular dystrophy. It has been demonstrated previously that repeated intravascular injection of phosphorodiamidate morpholino oligomers (PMOs) in the mdx mouse induces more dystrophin expression than a single injection, but this approach is costly, and data demonstrating the safety of high doses of systemically injected PMO are unavailable. Furthermore, several publications have demonstrated the efficacy of peptide-conjugated PMOs, but the clinical applicability of such compounds is unclear at this stage. Here, we report that multiple intravascular injections of low doses of naked PMO show significantly more dystrophin-positive fibers in a variety of muscle groups, 8 weeks after administration compared with a single dose of the same total amount. After administration of a total of 200 mg of PMO per kilogram, histological features, such as the cross-sectional area, centronucleation index, and expression of the dystrophin-associated protein complex, showed significant improvement in mice treated by repeated injection. Furthermore, four administrations of just 5 mg/kg induced a significant amount of dystrophin expression. These results clearly demonstrate the key role of the optimization of dosing regimen for the systemic administration of PMO in patients, and support the clinical feasibility of this approach with naked PMO.


Assuntos
Éxons/genética , Morfolinas/administração & dosagem , Distrofia Muscular de Duchenne/terapia , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Distrofina/metabolismo , Complexo de Proteínas Associadas Distrofina/genética , Complexo de Proteínas Associadas Distrofina/metabolismo , Terapia Genética/métodos , Humanos , Injeções Intramusculares , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos mdx , Morfolinas/metabolismo , Morfolinos , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/genética , Oligonucleotídeos Antissenso/administração & dosagem , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Resultado do Tratamento
12.
Mol Ther ; 17(3): 554-61, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19142179

RESUMO

Duchenne muscular dystrophy (DMD) is caused by out-of-frame mutations of the human DMD gene. Antisense oligonucleotides (AOs) have previously been used to skip additional exons that border the deletions such that the reading frame is restored and internally truncated, but functional, dystrophin expressed. We have designed phosphorodiamidate morpholino oligomer (PMO) AOs to various exons of the human dystrophin gene. PMOs were designed to have their target sites overlapping areas of open RNA structure, as defined by hybridization-array analysis, and likely exonic splicing enhancer (ESE)/silencer sites on the target RNA. The ability of each PMO to produce exon skipping was tested in vitro in normal human skeletal muscle cells. Retrospective analysis of design parameters used and PMO variables revealed that active PMOs were longer, bound to their targets more strongly, had their target sites closer to the acceptor splice site of the exon, overlapped areas of open conformation (as defined by the hybridization or the RNA secondary structure prediction software), and could interfere with the binding of certain SR proteins. No other parameter appeared to show significant association to PMO-skipping efficacy. No design tool is strong enough in isolation; however, if used in conjunction with other significant parameters it can aid AO design.


Assuntos
Desenho de Fármacos , Distrofina/genética , Distrofina/metabolismo , Éxons/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Morfolinas/farmacologia , Sequência de Bases , Células Cultivadas , Simulação por Computador , Distrofina/química , Humanos , Dados de Sequência Molecular , Morfolinos , Conformação de Ácido Nucleico
13.
Atherosclerosis ; 204(1): 121-6, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18930458

RESUMO

Hepatic gene transfer of atheroprotective human apoE by recombinant viral vectors can reverse hypercholesterolaemia and inhibit atherogenesis in apoE-deficient (apoE(-/-)) mice. Here, in preliminary studies we assess the effectiveness of a recently developed self-complementary adeno-associated virus (scAAV) serotype 8 vector, driven by a hepatocyte-specific promoter (LP1), for liver-directed gene delivery of human apoE3. Vector viability was validated by transducing cultured HepG2 cells and measuring secretion of apoE3 protein. Male and female apoE(-/-) mice, 6-month old and fed on normal chow, were intravenously injected with 1x10(11) vg (vector genomes) of scAAV2/8.LP1.apoE3; age-matched untreated mice served as controls. In male mice, plasma apoE3 levels were sufficiently high (up to 17 microg/ml) to normalize plasma total cholesterol and ameliorate their proatherogenic lipoprotein profile, by reducing VLDL/LDL and increasing HDL 5-fold. At termination (12 weeks) development of aortic atherosclerosis was significantly retarded by 58% (aortic lesion area 8.2+/-1.4% vs. 19.3+/-2.4% in control males; P<0.001). Qualitatively similar anti-atherogenic effects were noted when female mice were treated, but the benefits were less marked and aortic lesions, for example, were reduced by only 33% (15.7+/-3.7% vs. 23.6+/-6.9%). Although group numbers were small (n=4/5), this gender-specific difference reflected two to three times less apoE3 in plasma of female mice at weeks 3 and 6, implying that gene transfer to female liver using scAAV vectors may require additional optimization, despite their established superior potency to conventional single-stranded (ssAAV) vectors.


Assuntos
Apolipoproteína E3/biossíntese , Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos , Fígado/metabolismo , Transdução Genética , Animais , Aorta/metabolismo , Aorta/patologia , Apolipoproteína E3/sangue , Apolipoproteína E3/genética , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Linhagem Celular Tumoral , Colesterol/sangue , Modelos Animais de Doenças , Feminino , Humanos , Lipoproteínas HDL/sangue , Lipoproteínas LDL/sangue , Lipoproteínas VLDL/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Projetos Piloto , Regiões Promotoras Genéticas , Fatores Sexuais , Fatores de Tempo
14.
Hum Gene Ther ; 19(10): 1000-8, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18788904

RESUMO

Chromosomal correction of dystrophin gene mutations is a most desirable therapeutic solution for Duchenne muscular dystrophy, as it allows production of the full-length dystrophin under the control of locus-specific promoters. Here we explored gene targeting in conditionally immortal mouse dystrophin-deficient myoblasts. We constructed an adenoviral vector for the correction of the mdx mutation, containing 6.0 kb of sequence homologous to the target locus (partial intron 21 through to exon 24 with the normal sequence of exon 23) and a neomycin expression cassette inserted in intron 23. Adenovirus-based gene targeting was previously reported to be beneficial in mouse embryonic stem cells, resulting in one targeted integration per three integration events. However, we found no targeted integration events among 144 stably transduced G418-resistant myoblast clones, reflecting efficient random integration of the adenoviral vector in myogenic cells. We found that mouse myoblasts are capable of integrating recombinant adenoviral DNA with an efficiency approaching 1%. Interestingly, dermal fibroblasts integrate adenoviral DNA up to 100 times less efficiently than myoblasts from the same mice. We also show that the efficiency of recombinant adenoviral DNA integration is influenced by preinfection cell density, possibly indicating the importance of cellular DNA replication for adenoviral integration.


Assuntos
Adenoviridae , Distrofina , Marcação de Genes , Terapia Genética , Vetores Genéticos , Distrofia Muscular de Duchenne/terapia , Integração Viral , Animais , Replicação do DNA , Derme , Células-Tronco Embrionárias/metabolismo , Fibroblastos , Humanos , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Mutação , Mioblastos , Locos de Características Quantitativas/genética
15.
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
16.
Hum Gene Ther ; 19(6): 569-78, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18578629

RESUMO

Plasma apolipoprotein E (apoE) has multiple atheroprotective actions. However, although liver-directed adenoviral gene transfer of apoE reverses hypercholesterolemia and inhibits atherogenesis in apoE-deficient (apoE(-/-)) mice, safety considerations have revived interest in nonviral DNA (plasmid) and nonpathogenic adeno-associated viral (AAV) vectors. Here, we assess the effectiveness of these two delivery vehicles by minimally invasive intramuscular injection. First, we constructed AAV2-based expression plasmids harboring human apoE3 cDNA, driven by two muscle-specific promoters (CK6 and C5-12) and one ubiquitous promoter (CAG); each efficiently expressed apoE3 in transfected cultured C2C12 mouse myoblasts, although muscle-specific promoters were active only in differentiated multinucleate myotubes. Second, a pilot study verified that electrotransfer of the CAG-driven plasmid (p.CAG.apoE3) into tibialis anterior muscles, pretreated with hyaluronidase, of apoE(-/-) mice significantly enhanced (p < 0.001) local intramuscular expression of apoE3. However, in a 7-day experiment, the CK6- and C5-12-driven plasmids produced less apoE3 in muscle than did p.CAG.apoE3 (0.61 +/- 0.38 and 0.45 +/- 0.38 vs. 13.38 +/- 7.46 microg of apoE3 per muscle, respectively), but plasma apoE3 levels were below our detection limit (<15 ng/ml) in all mice and did not reverse the hyperlipidemia. Finally, we showed that intramuscular injection of a cross-packaged AAV serotype 7 viral vector, expressing human apoE3 from the CAG promoter, resulted in increasing levels of apoE3 in plasma over 4 weeks, although the concentration reached (1.40 +/- 0.35 microg/ml) was just below the threshold level needed to reduce the hypercholesterolemia. We conclude that skeletal muscle can serve as an effective secretory platform to express the apoE3 transgene, but that improved gene transfer vectors are needed to achieve full therapeutic levels of plasma apoE3 protein.


Assuntos
Apolipoproteínas E/biossíntese , Dependovirus , Eletroporação/métodos , Terapia Genética , Vetores Genéticos/administração & dosagem , Músculo Esquelético/metabolismo , Plasmídeos/administração & dosagem , Animais , Apolipoproteínas E/sangue , Apolipoproteínas E/genética , Aterosclerose/terapia , Células Cultivadas , DNA/administração & dosagem , DNA/genética , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Humanos , Injeções Intramusculares , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/citologia , Mioblastos/metabolismo , Plasmídeos/genética , Regiões Promotoras Genéticas , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/sangue , Proteínas Recombinantes/genética
17.
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
18.
Biochim Biophys Acta ; 1686(3): 190-9, 2005 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-15629688

RESUMO

Plasma apolipoprotein E (apoE) is a 34-kDa polymorphic protein which has atheroprotective actions by clearing remnant lipoproteins and sequestering excess cellular cholesterol. Low or dysfunctional apoE is a risk factor for hyperlipidaemia and atherosclerosis, and for restenosis after angioplasty. Here, in short-term studies designed to establish proof-of-principle, we investigate whether encapsulated recombinant Chinese hamster ovary (CHO) cells can secrete wild-type apoE3 protein in vitro and then determine whether peritoneal implantation of the microcapsules into apoE-deficient (apoE(-/-)) mice reduces their hypercholesterolaemia. Recombinant CHO-E3 cells were encapsulated into either alginate poly-l-lysine or alginate polyethyleneimine/polybrene microspheres. After verifying stability and apoE3 secretion, the beads were then implanted into the peritoneal cavity of apoE(-/-) mice; levels of plasma apoE3, cholesterol and lipoproteins were monitored for up to 14 days post-implantation. Encapsulated CHO-E3 cells continued to secrete apoE3 protein throughout a 60-day study period in vitro, though levels declined after 14 days. This cell-derived apoE3 was biologically active. When conditioned medium from encapsulated CHO-E3 cells was incubated with cultured cells pre-labelled with [(3)H]-cholesterol, efflux of cholesterol was two to four times greater than with normal medium (at 8 h, for example, 7.4+/-0.3% vs. 2.4+/-0.2% of cellular cholesterol; P<0.001). Moreover, when secreted apoE3 was injected intraperitoneally into apoE(-/-) mice, apoE3 was detected in plasma and the hyperlipidaemia improved. Similarly, when alginate polyethyleneimine/polybrene capsules were implanted into the peritoneum of apoE(-/-) mice, apoE3 was secreted into plasma and at 7 days total cholesterol was reduced, while atheroprotective high-density lipoprotein (HDL) increased. In a second study, apoE was detectable in plasma of five mice treated with alginate poly-l-lysine beads, 4 and 7 days post-implantation, though not at day 14. Furthermore, their hypercholesterolaemia was reduced, while HDL was clearly elevated in all mice at days 4 and 7 (from 18.4+/-6.2% of total lipoproteins to 31.1+/-6.8% at 7 days; P<0.001); however, these had rebounded by day 14, possibly due to the emergence of anti-apoE antibodies. We conclude that microencapsulated apoE-secreting cells have the potential to ameliorate the hyperlipidaemia of apoE deficiency, but that the technology must be improved to become a feasible therapeutic to treat atherosclerosis.


Assuntos
Apolipoproteínas E/genética , Células CHO , Transplante de Células/métodos , Hiperlipidemias/terapia , Alginatos/química , Animais , Apolipoproteínas E/metabolismo , HDL-Colesterol/sangue , Cricetinae , Cricetulus , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Injeções Intraperitoneais , Camundongos , Camundongos Knockout , Microesferas , Peritônio
19.
J Gene Med ; 6(10): 1149-58, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15386737

RESUMO

BACKGROUND: The activity of synthetic antisense oligonucleotides (splicomers) designed to block pre-mRNA splicing at specific exons has been demonstrated in a number of model systems, including constitutively spliced exons in mouse dystrophin RNA. Splicomer reagents directed to Duchenne muscular dystrophy (DMD) RNAs might thus circumvent nonsense or frame-shifting mutations, leading to therapeutic expression of partially functional dystrophin, as occurs in the milder, allelic (Becker) form of the disease (BMD). METHODS: Functional and hybridisation array screens have been used to select optimised splicomers directed to exon 23 of dystrophin mRNA which carries a nonsense mutation in the mdx mouse. Splicomers were transfected into cultured primary muscle cells, and dystrophin mRNA assessed for exon exclusion. Splicomers were also administered to the muscles of mdx mice. RESULTS: Oligonucleotide array analyses with dystrophin pre-mRNA probes revealed strong and highly specific hybridisation patterns spanning the exon 23/intron 23 boundary, indicating an open secondary structure conformation in this region of the RNA. Functional screening of splicomer arrays by direct analysis of exon 23 RNA splicing in mdx muscle cultures identified a subset of biologically active reagents which target sequence elements associated with the 5' splice site region of dystrophin intron 23; splicomer-mediated exclusion of exon 23 was specific and dose-responsive up to a level exceeding 50% of dystrophin mRNA, and Western blotting demonstrated de novo expression of dystrophin protein at 2-5% of wild-type levels. Direct intramuscular administration of optimised splicomer reagents in vivo resulted in the reappearance of sarcolemmal dystrophin immunoreactivity in > 30% of muscle fibres in the mdx mouse CONCLUSIONS: These results suggest that correctly designed splicomers may have direct therapeutic value in vivo, not only for DMD, but also for a range of other genetic disorders.


Assuntos
Distrofina/genética , Músculos/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Alelos , Processamento Alternativo , Animais , Sequência de Bases , Western Blotting , DNA/genética , DNA/metabolismo , Relação Dose-Resposta a Droga , Distrofina/biossíntese , Éxons , Mutação da Fase de Leitura , Terapia Genética , Imuno-Histoquímica , Íntrons , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Dados de Sequência Molecular , Músculos/citologia , Distrofia Muscular de Duchenne/terapia , Hibridização de Ácido Nucleico , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos Antissenso/genética , Ligação Proteica , RNA/metabolismo , Splicing de RNA , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
20.
Hum Mol Genet ; 11(15): 1719-30, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12095914

RESUMO

The ability to transfer the dystrophin gene stably to the skeletal muscle of DMD patients is a major confounding issue in establishing an effective gene therapy for this disease. To overcome this problem, we have examined the ability of muscle fibres from mdx mice to act as in situ factories of retroviral vector production. Tibialis anterior (TA) muscles from 4-week-old mdx mice were injected with an adenoviral vector expressing LacZ within a retroviral expression cassette (AdLZIN). Retroviral vector production was induced by the inclusion of two additional adenoviral vectors expressing retroviral gag-pol (AdGagPol) and 10A1 env genes (Ad10A1). Upon introduction of infected muscles into cell culture, colonies of beta-galactosidase-expressing myotubes formed only in cultures where the muscle was injected with AdLZIN, AdGagPol and Ad10A1, but not from muscle injected with AdLZIN only. Muscles from mdx/nude mice producing retroviral vector displayed a 4.6-fold increase in beta-galactosidase-positive myofibres after 1 month, compared with contralateral muscle in the same animal injected with AdLZIN and AdGagPol only. By constructing a hybrid adeno-retroviral vector expressing a truncated micro-dystrophin construct (AdmicroDyIN), we were able to partially correct the mdx dystrophic phenotype. AdmicroDyIN-mediated expression of micro-dystrophin in mdx TA muscle restored the formation of the dystrophin-associated glycoprotein complex and significantly reduced the level of muscle degeneration over uninjected controls. By stimulating in situ production of retroviral vector expressing micro-dystrophin, we achieved 92%+/-6% transduction of myofibres in the TA muscle by 4 weeks. Strikingly, by 3 months post injection, micro-dystrophin was still expressed to high levels in nearly all the myofibres of the TA muscle. By comparison, there was a pronounced drop in the levels of micro-dystrophin expressed by muscles injected with AdmicroDyIN only. Finally, using a novel PCR approach, we detected reverse-transcribed, integrated proviral sequences in TA muscle genomic DNA by 4 weeks post injection, the levels of which were found to increase after 3 months.


Assuntos
Adenoviridae/genética , Distrofina/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Distrofia Muscular de Duchenne/terapia , Retroviridae/genética , Animais , Modelos Animais de Doenças , Terapia Genética , Óperon Lac , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/fisiopatologia , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...