Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hum Gene Ther ; 31(21-22): 1155-1168, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32940068

RESUMO

Recently, we established an adeno-associated virus (AAV9) capsid-promoter interaction that directly determined cell-specific gene expression across two synthetic promoters, Cbh and CBA, in the rat striatum. These studies not only expand this capsid-promoter interaction to include another promoter in the rat striatum but also establish AAV capsid-promoter interactions in the nonhuman primate brain. When AAV serotype 9 (AAV9) vectors were injected into the rat striatum, the minimal synthetic promoter JetI drove green fluorescent protein (GFP) gene expression predominantly in oligodendrocytes. However, similar to our previous findings, the insertion of six alanines into VP1/VP2 of the AAV9 capsid (AAV9AU) significantly shifted JetI-driven GFP gene expression to neurons. In addition, previous retrograde tracing studies in the nonhuman primate brain also revealed the existence of a capsid-promoter interaction. When rAAV2-Retro vectors were infused into the frontal eye field (FEF) of rhesus macaques, local gene expression was prominent using either the hybrid chicken beta actin (CAG) or human synapsin (hSyn) promoters. However, only the CAG promoter, not the hSyn promoter, led to gene expression in the ipsilateral claustrum and contralateral FEF. Conversely, infusion of rAAV2-retro-hSyn vectors, but not rAAV2-retro-CAG, into the macaque superior colliculus led to differential and selective retrograde gene expression in cerebellotectal afferent cells. Clearly, this differential promoter/capsid expression profile could not be attributed to promoter inactivation from retrograde transport of the rAAV2-Retro vector. In summary, we document the potential for AAV capsid/promoter interactions to impact cell-specific gene expression across species, experimental manipulations, and engineered capsids, independent of capsid permissivity.


Assuntos
Encéfalo/metabolismo , Capsídeo/metabolismo , Dependovirus/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Regiões Promotoras Genéticas , Transgenes , Animais , Dependovirus/genética , Macaca mulatta , Masculino , Ratos , Ratos Sprague-Dawley
2.
Mol Ther ; 28(5): 1373-1380, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32213322

RESUMO

Cell-selective gene expression comprises a critical element of many adeno-associated virus (AAV) vector-based gene therapies, and to date achieving this goal has focused on AAV capsid engineering, cell-specific promoters, or cell-specific enhancers. Recently, we discovered that the capsid of AAV9 exerts a differential influence on constitutive promoters of sufficient magnitude to alter cell type gene expression in the rat CNS. For AAV9 vectors chicken ß-actin (CBA) promoter-driven gene expression exhibited a dominant neuronal gene expression in the rat striatum. Surprisingly, for otherwise identical AAV9 vectors, the truncated CBA hybrid (CBh) promoter shifted gene expression toward striatal oligodendrocytes. In contrast, AAV2 vector gene expression was restricted to striatal neurons, regardless of the constitutive promoter used. Furthermore, a six-glutamate residue insertion immediately after the VP2 start residue shifted CBA-driven cellular gene expression from neurons to oligodendrocytes. Conversely, a six-alanine insertion in the same AAV9 capsid region reversed the CBh-mediated oligodendrocyte expression back to neurons without changing AAV9 capsid access to oligodendrocytes. Given the preponderance of AAV9 in ongoing clinical trials and AAV capsid engineering, this AAV9 capsid-promoter interaction reveals a previously unknown novel contribution to cell-selective AAV-mediated gene expression in the CNS.


Assuntos
Proteínas do Capsídeo/genética , Capsídeo/metabolismo , Corpo Estriado/metabolismo , Dependovirus/genética , Expressão Gênica , Neurônios/metabolismo , Regiões Promotoras Genéticas , Animais , Terapia Genética , Vetores Genéticos , Células HEK293 , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Transdução Genética , Transgenes
3.
Mol Ther ; 25(4): 928-934, 2017 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-28202388

RESUMO

Recent advances suggest that in vivo reprogramming of endogenous cell populations provides a viable alternative for neuron replacement. Astrocytes and oligodendrocyte precursor cells can be induced to transdifferentiate into neurons in the CNS, but, in these instances, reprogramming requires either transgenic mice or retroviral-mediated gene expression. We developed a microRNA (miRNA)-GFP construct that in vitro significantly reduced the expression of polypyrimidine tract-binding protein, and, subsequently, we packaged this construct in a novel oligodendrocyte preferring adeno-associated virus vector. Ten days after rat striatal transduction, the vast majority of the GFP-positive cells were oligodendrocytes, but 6 weeks to 6 months later, the majority of GFP-positive cells exhibited neuronal morphology and co-localized with the neuronal marker NeuN. Patch-clamp studies on striatal slices established that the GFP-positive cells exhibited electrophysiological properties indicative of mature neurons, such as spontaneous action potentials and spontaneous inhibitory postsynaptic currents. Also, 3 months after striatal vector administration, GFP-positive terminals in the ipsilateral globus pallidus or substantia nigra retrogradely transported fluorescent beads back to GFP-positive striatal cell bodies, indicating the presence of functional presynaptic terminals. Thus, this viral vector approach provides a potential means to harness resident oligodendrocytes as an endogenous source for in vivo neuronal replacement.


Assuntos
Transdiferenciação Celular/genética , Reprogramação Celular/genética , Corpo Estriado/citologia , Vetores Genéticos/genética , Neurônios/citologia , Oligodendroglia/citologia , Animais , Linhagem Celular , Dependovirus/genética , Humanos , Neurônios/metabolismo , Oligodendroglia/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Ratos
4.
Neurobiol Dis ; 96: 323-334, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27717881

RESUMO

Breakdown of neuro-glial N-acetyl-aspartate (NAA) metabolism results in the failure of developmental myelination, manifest in the congenital pediatric leukodystrophy Canavan disease caused by mutations to the sole NAA catabolizing enzyme aspartoacylase. Canavan disease is a major point of focus for efforts to define NAA function, with available evidence suggesting NAA serves as an acetyl donor for fatty acid synthesis during myelination. Elevated NAA is a diagnostic hallmark of Canavan disease, which contrasts with a broad spectrum of alternative neurodegenerative contexts in which levels of NAA are inversely proportional to pathological progression. Recently generated data in the nur7 mouse model of Canavan disease suggests loss of aspartoacylase function results in compromised energetic integrity prior to oligodendrocyte death, abnormalities in myelin content, spongiform degeneration, and motor deficit. The present study utilized a next-generation "oligotropic" adeno-associated virus vector (AAV-Olig001) to quantitatively assess the impact of aspartoacylase reconstitution on developmental myelination. AAV-Olig001-aspartoacylase promoted normalization of NAA, increased bioavailable acetyl-CoA, and restored energetic balance within a window of postnatal development preceding gross histopathology and deteriorating motor function. Long-term effects included increased oligodendrocyte numbers, a global increase in myelination, reversal of vacuolation, and rescue of motor function. Effects on brain energy observed following AAV-Olig001-aspartoacylase gene therapy are shown to be consistent with a metabolic profile observed in mild cases of Canavan disease, implicating NAA in the maintenance of energetic integrity during myelination via oligodendroglial aspartoacylase.


Assuntos
Amidoidrolases/metabolismo , Ácido Aspártico/análogos & derivados , Encéfalo/enzimologia , Doença de Canavan/patologia , Bainha de Mielina/fisiologia , Oligodendroglia/enzimologia , Amidoidrolases/genética , Animais , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Proteínas Relacionadas à Autofagia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Encéfalo/patologia , Doença de Canavan/complicações , Doença de Canavan/diagnóstico por imagem , Doença de Canavan/genética , Criança , Pré-Escolar , Dependovirus/genética , Progressão da Doença , Metabolismo Energético/genética , Feminino , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Lactente , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Transtornos dos Movimentos/etiologia , Proteína Básica da Mielina/metabolismo , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/genética
5.
J Neurosci ; 34(17): 5824-34, 2014 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-24760842

RESUMO

Neuroactive steroids are endogenous neuromodulators capable of altering neuronal activity and behavior. In rodents, systemic administration of endogenous or synthetic neuroactive steroids reduces ethanol self-administration. We hypothesized this effect arises from actions within mesolimbic brain regions that we targeted by viral gene delivery. Cytochrome P450 side chain cleavage (P450scc) converts cholesterol to pregnenolone, the rate-limiting enzymatic reaction in neurosteroidogenesis. Therefore, we constructed a recombinant adeno-associated serotype 2 viral vector (rAAV2), which drives P450scc expression and neuroactive steroid synthesis. The P450scc-expressing vector (rAAV2-P450scc) or control GFP-expressing vector (rAAV2-GFP) were injected bilaterally into the ventral tegmental area (VTA) or nucleus accumbens (NAc) of alcohol preferring (P) rats trained to self-administer ethanol. P450scc overexpression in the VTA significantly reduced ethanol self-administration by 20% over the 3 week test period. P450scc overexpression in the NAc, however, did not alter ethanol self-administration. Locomotor activity was unaltered by vector administration to either region. P450scc overexpression produced a 36% increase in (3α,5α)-3-hydroxypregnan-20-one (3α,5α-THP, allopregnanolone)-positive cells in the VTA, but did not increase 3α,5α-THP immunoreactivity in NAc. These results suggest that P450scc overexpression and the resultant increase of 3α,5α-THP-positive cells in the VTA reduces ethanol reinforcement. 3α,5α-THP is localized to neurons in the VTA, including tyrosine hydroxylase neurons, but not astrocytes. Overall, the results demonstrate that using gene delivery to modulate neuroactive steroids shows promise for examining the neuronal mechanisms of moderate ethanol drinking, which could be extended to other behavioral paradigms and neuropsychiatric pathology.


Assuntos
Álcoois/administração & dosagem , Enzima de Clivagem da Cadeia Lateral do Colesterol/metabolismo , Condicionamento Operante/fisiologia , Etanol/administração & dosagem , Pregnanolona/metabolismo , Área Tegmentar Ventral/metabolismo , Animais , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Condicionamento Operante/efeitos dos fármacos , Masculino , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Ratos , Ratos Wistar , Autoadministração , Área Tegmentar Ventral/efeitos dos fármacos
6.
Exp Neurol ; 247: 429-37, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23333565

RESUMO

Resected epileptic tissues exhibit elements of chronic neuroinflammation that include elevated TNFα and increased TNFα receptor activation, but the seizure related consequences of chronic TNFα expression remain unknown. Twenty four hours after acute limbic seizures the rat hippocampus exhibited a rapid upregulation of TNFR1, but a simultaneous downregulation of TNFR2. These limbic seizures also evoked significant increases in measures of neuroinflammation and caused significant neuronal cell death in both the hilus and CA3 of the hippocampus. In order to mimic a state of chronic TNFα exposure, adeno-associated viral vectors were packaged with a TNF receptor 1 (TNFR1) specific agonist, human TNFα, or a TNF receptor 1/2 agonist, rat TNFα. Subsequently, chronic hippocampal overexpression of either TNFR ligand caused microglial activation and blood-brain barrier compromise, a pattern similar to limbic seizure-induced neuroinflammation. However, no evidence was found for neuronal cell death or spontaneous seizure activity. Thus, chronic, in vivo TNFα expression and the subsequent neuroinflammation alone did not cause cell death or elicit seizure activity. In contrast, chronic hippocampal activation of TNFR1 alone significantly increased limbic seizure sensitivity in both amygdala kainic acid and electrical amygdala kindling models, while chronic activation of both TNFR1 and TNFR2 significantly attenuated the amygdala kindling rate. With regard to endogenous TNFα, chronic hippocampal expression of a TNFα decoy receptor significantly reduced seizure-induced cell death in the hippocampus, but did not alter seizure susceptibility. These findings suggest that blockade of endogenous TNFα could attenuate seizure related neuropathology, while selective activation of TNFR2 could exert beneficial therapeutic effects on in vivo seizure sensitivity.


Assuntos
Hipocampo/metabolismo , Sistema Límbico/patologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Convulsões/patologia , Fator de Necrose Tumoral alfa/metabolismo , Análise de Variância , Animais , Antígeno CD11b , Linhagem Celular Transformada , Regulação da Expressão Gênica/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Excitação Neurológica , Masculino , Proteínas do Tecido Nervoso/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fatores de Tempo , Transfecção , Fator de Necrose Tumoral alfa/biossíntese
7.
Neuropharmacology ; 69: 82-8, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22465202

RESUMO

Diseases of the central nervous system (CNS) have provided enormous opportunities for the therapeutic application of viral vector gene transfer. Adeno-associated virus (AAV) has been the vector of choice in recent clinical trials of neurological disease, including Parkinson's and Alzheimer's disease, due to the safety, efficacy, and stability of AAV gene transfer to the CNS. This review highlights the strategies employed for improving direct and peripheral targeting of therapeutic vectors to CNS tissue, and considers the significance of cellular and tissue transduction specificity, transgene regulation, and other variables that influence achievement of successful therapeutic goals. This article is part of the Special Issue entitled 'New Targets and Approaches to the Treatment of Epilepsy'.


Assuntos
Dependovirus/genética , Terapia Genética , Doenças do Sistema Nervoso/terapia , Animais , Expressão Gênica , Vetores Genéticos , Humanos , Injeções Intraventriculares , Transgenes/genética
8.
Exp Neurol ; 244: 27-35, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22008258

RESUMO

This review addresses the state of gene therapy research for the treatment of epilepsy. Preclinical studies have demonstrated the anti-seizure efficacy of viral vector-based gene transfer through the use of a variety of strategies - from modulating classic neurotransmitter systems to targeting or overexpressing of neuropeptide receptors in seizure-specific brain regions. While these studies provide substantive proof of principle for viral vector gene therapy, future studies must address the challenges of vector immunity, cellular specificity and effective global delivery. As these issues are resolved, viral vector gene therapy should significantly impact the treatment of intractable epilepsy.


Assuntos
Epilepsia/terapia , Terapia Genética/métodos , Animais , Epilepsia/genética , Técnicas de Transferência de Genes , Vetores Genéticos/fisiologia , Humanos
9.
Addict Biol ; 17(2): 338-50, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21762289

RESUMO

Neuropeptide Y (NPY) and protein kinase A (PKA) have been implicated in neurobiological responses to ethanol. We have previously reported that mutant mice lacking normal production of the RIIß subunit of PKA (RIIß-/- mice) show enhanced sensitivity to the locomotor stimulant effects of ethanol and increased behavioral sensitization relative to littermate wild-type RIIß+/+ mice. We now report that RIIß-/- mice also show increased NPY immunoreactivity in the nucleus accumbens (NAc) core and the ventral striatum relative to RIIß+/+ mice. These observations suggest that elevated NPY signaling in the NAc and/or striatum may contribute to the increased sensitivity to ethanol-induced behavioral sensitization that is a characteristic of RIIß-/- mice. Consistently, NPY-/- mice failed to display ethanol-induced behavioral sensitization that was evident in littermate NPY+/+ mice. To examine more directly the role of NPY in the locomotor stimulant effects of ethanol, we infused a recombinant adeno-associated virus (rAAV) into the region of the NAc core of DBA/2J mice. The rAAV-fibronectin (FIB)-NPY(13-36) vector expresses and constitutively secretes the NPY fragment NPY(13-36) (a selective Y(2) receptor agonist) from infected cells in vivo. Mice treated with the rAAV-FIB-NPY(13-36) vector exhibited reduced expression of ethanol-induced behavioral sensitization compared with mice treated with a control vector. Taken together, the current data provide the first evidence that NPY signaling in the NAc core and the Y(2) receptor modulate ethanol-induced behavioral sensitization.


Assuntos
Gânglios da Base/metabolismo , Depressores do Sistema Nervoso Central/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Etanol/farmacologia , Neuropeptídeo Y/metabolismo , Núcleo Accumbens/metabolismo , Consumo de Bebidas Alcoólicas/metabolismo , Consumo de Bebidas Alcoólicas/psicologia , Animais , Comportamento Animal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos , Atividade Motora/efeitos dos fármacos , Neuropeptídeo Y/farmacologia , Fragmentos de Peptídeos/farmacologia , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Reforço Psicológico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia
10.
Curr Protoc Neurosci ; Chapter 4: Unit 4.17, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21971848

RESUMO

Adeno-associated virus is a nonpathogenic human virus that has been developed into a gene-delivery vector due to its high efficiency of infection for many different cell types and its ability to persist and lead to long-term gene expression. This unit describes efficient methods to generate high-titer, research-grade, adenovirus-free recombinant single-stranded and self-complementary adeno-associated virus in various serotypes, along with methods to quantify the viral vectors. Two detailed methods are provided for viral vector delivery into the rodent brain and spinal cord, and for histological detection of transgene expression of GFP.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Animais , Encéfalo/metabolismo , Dependovirus/metabolismo , Vetores Genéticos/administração & dosagem
11.
Curr Gene Ther ; 11(3): 181-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21453285

RESUMO

Adeno-associated virus (AAV) vectors exhibit a number of properties that have made this vector system an excellent choice for both CNS gene therapy and basic neurobiological investigations. In vivo, the preponderance of AAV vector transduction occurs in neurons where it is possible to obtain long-term, stable gene expression with very little accompanying toxicity. Promoter selection, however, significantly influences the pattern and longevity of neuronal transduction distinct from the tropism inherent to AAV vectors. AAV vectors have successfully manipulated CNS function using a wide variety of approaches including expression of foreign genes, expression of endogenous genes, expression of antisense RNA and expression of RNAi. With the discovery and characterization of different AAV serotypes, as well as the creation of novel chimeric serotypes, the potential patterns of in vivo vector transduction have been expanded substantially, offering alternatives to the more studied AAV 2 serotype. Furthermore, the development of specific AAV chimeras offers the potential to further refine targeting strategies. These different AAV serotypes also provide a solution to the immune silencing that proves to be a realistic likelihood given broad exposure of the human population to the AAV 2 serotype. These advantageous CNS properties of AAV vectors have fostered a wide range of clinically relevant applications including Parkinson's disease, lysosomal storage diseases, Canavan's disease, epilepsy, Huntington's disease and ALS. In many cases the proposed therapies have progressed to phase I/II clinical trials. Each individual application, however, presents a unique set of challenges that must be solved in order to attain clinically effective gene therapies.


Assuntos
Doenças do Sistema Nervoso Central/terapia , Sistema Nervoso Central/virologia , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos , Animais , Humanos
12.
Hum Gene Ther ; 22(9): 1143-53, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21476867

RESUMO

With the increased use of small self-complementary adeno-associated viral (AAV) vectors, the design of compact promoters becomes critical for packaging and expressing larger transgenes under ubiquitous or cell-specific control. In a comparative study of commonly used 800-bp cytomegalovirus (CMV) and chicken ß-actin (CBA) promoters, we report significant differences in the patterns of cell-specific gene expression in the central and peripheral nervous systems. The CMV promoter provides high initial neural expression that diminishes over time. The CBA promoter displayed mostly ubiquitous and high neural expression, but substantially lower expression in motor neurons (MNs). We report the creation of a novel hybrid form of the CBA promoter (CBh) that provides robust long-term expression in all cells observed with CMV or CBA, including MNs. To develop a short neuronal promoter to package larger transgenes into AAV vectors, we also found that a 229-bp fragment of the mouse methyl-CpG-binding protein-2 (MeCP2) promoter was able to drive neuron-specific expression within the CNS. Thus the 800-bp CBh promoter provides strong, long-term, and ubiquitous CNS expression whereas the MeCP2 promoter allows an extra 570-bp packaging capacity, with low and mostly neuronal expression within the CNS, similar to the MeCP2 transcription factor.


Assuntos
Sistema Nervoso Central/metabolismo , Dependovirus/genética , Regulação Viral da Expressão Gênica , Vetores Genéticos/genética , Sistema Nervoso Periférico/metabolismo , Regiões Promotoras Genéticas , Transdução Genética , Actinas/genética , Animais , Elementos Facilitadores Genéticos , Feminino , Perfilação da Expressão Gênica , Inativação Gênica , Células HEK293 , Humanos , Proteína 2 de Ligação a Metil-CpG/genética , Camundongos , Camundongos Endogâmicos BALB C , Neurônios/metabolismo , Especificidade de Órgãos/genética , Ratos , Ratos Sprague-Dawley
13.
Drug News Perspect ; 23(5): 281-6, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20603651

RESUMO

Adeno-associated virus (AAV) vectors support long-term, nontoxic gene expression in the central nervous system, and these AAV properties prove particularly applicable to the treatment of focal epilepsies, especially intractable temporal lobe epilepsy. A number of clinical studies have employed AAV vectors and to date, no known adverse effects have been directly associated with these treatments, particularly AAV serotype 2 (AAV2). Although other AAV serotypes may confer an advantage in the future, extensive studies on the inhibitory neuropeptides, galanin and neuropeptide Y, have generated enough preclinical evidence of efficacy to warrant AAV2-based clinical trials in the near future. Beyond these trials, emerging evidence suggests that AAV-mediated manipulation of adenosine can significantly impact limbic seizure activity. Thus, with appropriate nonhuman primate transduction patterns and favorable overall toxicology studies, AAV-based manipulation of adenosine could follow the AAV-neuropeptide clinical studies. Finally, recent findings using AAV capsid shuffling and directed evolution have identified a hybrid AAV vector that can selectively cross the seizure compromised blood-brain barrier and transduce cells after peripheral, intravenous administration. Thus, in the more distant future, AAV therapeutics for focal epilepsies may be delivered without any neurosurgical interventions.


Assuntos
Dependovirus/genética , Epilepsia/terapia , Terapia Genética , Vetores Genéticos , Adenosina/genética , Animais , Técnicas de Transferência de Genes , Terapia Genética/métodos , Humanos , Neuropeptídeos/genética , Neuropeptídeos/fisiologia
14.
J Virol ; 84(17): 8888-902, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20573820

RESUMO

The N termini of the capsid proteins VP1 and VP2 of adeno-associated virus (AAV) play important roles in subcellular steps of infection and contain motifs that are highly homologous to a phospholipase A(2) (PLA(2)) domain and nuclear localization signals (NLSs). To more clearly understand how virion components influence infection, we have generated mutations in these regions and examined their effects on subcellular trafficking, capsid stability, transduction, and sensitivity to pharmacological enhancement. All mutants tested assembled into capsids; retained the correct ratio of VP1, VP2, and VP3; packaged DNA similarly to recombinant AAV2 (rAAV2); and displayed similar stability profiles when heat denatured. Confocal microscopy demonstrated that these mutants trafficked through a perinuclear region in the vicinity of the Golgi apparatus, with a subset of mutants displaying more-diffuse localization consistent with an NLS-deficient phenotype. When tested for viral transduction, two mutant classes emerged. Class I (BR1(-), BR2(-), and BR2+K) displayed partial transduction, whereas class II (VP3 only, (75)HD/AN, BR3(-), and BR3+K) were severely defective. Surprisingly, one class II mutant (BR3+K) trafficked identically to rAAV2 and accumulated in the nucleolus, a step recently described by our laboratory that occurs with wild-type infection. The BR3+K mutant, containing an alanine-to-lysine substitution in the third basic region of VP1, was 10- to 100-fold-less infectious than rAAV2 in transformed cell lines (such as HEK-293, HeLa, and CV1-T cells), but in contrast, it was indistinguishable from rAAV2 in several nontransformed cell lines, as well as in tissues (liver, brain, and muscle) in vivo. Complementation studies with pharmacological adjuvants or adenovirus coinfection suggested that additional positive charges in NLS regions restrict mobilization in the nucleus and limit transduction in a transformed-cell-specific fashion. Remarkably, besides displaying cell-type-specific transduction, this is the first description of a capsid mutant indicating that nuclear entry is not sufficient for AAV-mediated transduction and suggests that additional steps (i.e., subnuclear mobilization or uncoating) limit successful AAV infection.


Assuntos
Substituição de Aminoácidos , Aminoácidos Básicos/química , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Dependovirus/fisiologia , Infecções por Parvoviridae/virologia , Transdução Genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Aminoácidos Básicos/genética , Aminoácidos Básicos/metabolismo , Animais , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Dependovirus/química , Dependovirus/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Mutação , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Alinhamento de Sequência , Replicação Viral
15.
Neuropsychopharmacology ; 35(4): 1016-25, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20032967

RESUMO

Excess neural activity in the CA3 region of the hippocampus has been linked to memory impairment in aged rats. We tested whether interventions aimed at reducing this excess activity would improve memory performance. Aged (24 to 28 months old) male Long-Evans rats were characterized in a spatial memory task known to depend on the functional integrity of the hippocampus, such that aged rats with identified memory impairment were used in a series of experiments. Overexpression of the inhibitory neuropeptide Y 13-36 in the CA3 via adeno-associated viral transduction was found to improve hippocampal-dependent long-term memory in aged rats, which had been characterized with impairment. Subsequent experiments with two commonly used antiepileptic agents, sodium valproate and levetiracetam, similarly produced dose-dependent memory improvement in such aged rats. Improved spatial memory with low doses of these agents was observed in both appetitve and aversive spatial tasks. The benefits of these different modalities of treatment are consistent with the concept that excess activity in the CA3 region of the hippocampus is a dysfunctional condition that may have a key role underlying age-related impairment in hippocampal-dependent memory processes. Because increased hippocampal activation occurs in age-related memory impairment in humans as observed in functional neuroimaging, the current findings also suggest that low doses of certain antiepileptic drugs in cognitively impaired elderly humans may have therapeutic potential and point to novel targets for this indication.


Assuntos
Envelhecimento , Região CA3 Hipocampal/fisiologia , Transtornos Cognitivos/patologia , Fatores Etários , Animais , Anticonvulsivantes/uso terapêutico , Região CA3 Hipocampal/efeitos dos fármacos , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Levetiracetam , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Neuropeptídeo Y/administração & dosagem , Neuropeptídeo Y/farmacologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/farmacologia , Piracetam/análogos & derivados , Piracetam/uso terapêutico , Ratos , Ratos Long-Evans , Percepção Espacial/efeitos dos fármacos , Percepção Espacial/fisiologia , Transdução Genética/métodos , Ácido Valproico/uso terapêutico
16.
Mol Ther ; 18(3): 570-8, 2010 03.
Artigo em Inglês | MEDLINE | ID: mdl-20040913

RESUMO

DNA shuffling and directed evolution were employed to develop a novel adeno-associated virus (AAV) vector capable of crossing the seizure-compromised blood-brain barrier (BBB) and transducing cells in the brain. Capsid DNA from AAV serotypes 1-6, 8, and 9 were shuffled and recombined to create a library of chimeric AAVs. One day after kainic acid-induced limbic seizure activity in rats, the virus library was infused intravenously (i.v.), and 3 days later, neuron-rich cells were mechanically dissociated from seizure-sensitive brain sites, collected and viral DNA extracted. After three cycles of selection, green fluorescent protein (GFP)-packaged clones were administered directly into brain or i.v. 1 day after kainic acid-induced seizures. Several clones that were effective after intracranial administration did not transduce brain cells after the i.v. administration. However, two clones (32 and 83) transduced the cells after direct brain infusion and after i.v. administration transduced the cells that were localized to the piriform cortex and ventral hippocampus, areas exhibiting a seizure-compromised BBB. No transduction occurred in areas devoid of BBB compromise. Only one parental serotype (AAV8) exhibited a similar expression profile, but the biodistribution of 32 and 83 diverged dramatically from this parental serotype. Thus, novel AAV vectors have been created that can selectively cross the seizure-compromised BBB and transduce cells.


Assuntos
Barreira Hematoencefálica , Dependovirus/metabolismo , Evolução Molecular Direcionada , Terapia Genética/métodos , Animais , Barreira Hematoencefálica/química , Capsídeo/química , Linhagem Celular , Sobrevivência Celular , DNA/metabolismo , Feminino , Proteínas de Fluorescência Verde/química , Humanos , Imuno-Histoquímica/métodos , Ácido Caínico/química , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal/métodos , Neurônios/metabolismo , Ratos , Convulsões/metabolismo
17.
Neurotherapeutics ; 6(2): 307-11, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19332324

RESUMO

Theoretically, gene therapy techniques offer an attractive alternative treatment option for intractable, focal epilepsies. Although logical gene therapy targets include excitatory and inhibitory receptors, variable viral vector tropism interjects an uncertainty as to the direction of change, seizure suppression, or seizure sensitization. To circumvent this therapeutic liability, adeno-associated virus (AAV) vectors have been constructed where the gene product is constitutively secreted from the transduced cell. Using AAV vectors, the fibronectin secretory signal sequence (FIB) was placed in front of the coding sequence for green fluorescent protein or the active portion of the neuroactive peptide galanin (GAL). Subsequent studies showed that these vectors supported expression and constitutive secretion of these gene products from transfected cells in vitro. More importantly, upon transduction in vivo, AAV-FIB-GAL vectors significantly attenuated focal seizure sensitivity, and this seizure attenuation could be controlled in vivo by using a tetracycline-regulated promoter. The expression and constitutive secretion of green fluorescent protein, or the expression of GAL alone, exerted no effect on focal seizure sensitivity. Moreover, unilateral infusion of the AAV-FIB-GAL vectors into the hippocampus prevented kainic acid-induced hilar cell death. With regard to limbic seizures, bilateral infusion of AAV-FIB-GAL vectors into the piriform cortex prevented both behavioral and localized electrographic seizure activity after the peripheral administration of kainic acid. Also, when rats were electrically kindled to class V seizure activity, subsequent infusion of AAV-FIB-GAL proved capable of significantly elevating the seizure initiation threshold. Thus, these studies clearly demonstrate the anti-seizure effectiveness of AAV vector-mediated expression and constitutive secretion of galanin.


Assuntos
Dependovirus/genética , Epilepsias Parciais/terapia , Galanina/metabolismo , Terapia Genética/métodos , Vetores Genéticos , Animais , Galanina/genética , Humanos , Ratos , Transdução Genética
18.
J Comp Neurol ; 511(2): 271-85, 2008 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-18785628

RESUMO

Rats lesioned with 6-hydroxydopamine (6-OHDA) as neonates exhibit behavioral and neurochemical abnormalities in adulthood that mimic Lesch-Nyhan disease, schizophrenia, and other developmental disorders of frontostriatal circuit dysfunction. In these animals a latent sensitivity to D1 agonists is maximally exposed by repeated administration of dopamine agonists in the postpubertal period (D1 priming). In neonate-lesioned, adult rats primed with SKF-38393, we found selective, persistent alterations in the morphology of pyramidal neuron apical dendrites in the prelimbic area of the medial prefrontal cortex (mPFC). In these animals, dendrite bundling patterns and the typically straight trajectories of primary dendritic shafts were disrupted, whereas the diameter of higher-order oblique branches was increased. Although not present in neonate-lesioned rats treated with saline, these morphological changes persisted at least 21 days after repeated dosing with SKF-38393, and were not accompanied by markers of neurodegenerative change. A sustained increase in phospho-ERK immunoreactivity in wavy dendritic shafts over the same period suggested a relationship between prolonged ERK phosphorylation and dendritic remodeling in D1-primed rats. In support of this hypothesis, pretreatment with the MEK1/2-ERK1/2 pathway inhibitors PD98059 or SL327, prior to each priming dose of SKF-38393, prevented the morphological changes associated with D1 priming. Together, these findings demonstrate that repeated stimulation of D1 receptors in adulthood interacts with the developmental loss of dopamine to profoundly and persistently modify neuronal signaling and dendrite morphology in the mature prefrontal cortex. Furthermore, sustained elevation of ERK activity in mPFC pyramidal neurons may play a role in guiding these morphological changes in vivo.


Assuntos
2,3,4,5-Tetra-Hidro-7,8-Di-Hidroxi-1-Fenil-1H-3-Benzazepina/metabolismo , Dendritos/ultraestrutura , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Córtex Pré-Frontal/anatomia & histologia , Córtex Pré-Frontal/metabolismo , Receptores de Dopamina D1/agonistas , Aminoacetonitrila/análogos & derivados , Aminoacetonitrila/metabolismo , Animais , Agonistas de Dopamina/metabolismo , Flavonoides/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Oxidopamina/toxicidade , Fosforilação , Córtex Pré-Frontal/efeitos dos fármacos , Inibidores de Proteases/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1/metabolismo , Simpatolíticos/toxicidade
19.
Mol Ther ; 16(7): 1252-60, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18500254

RESUMO

We report a DNA shuffling-based approach for developing cell type-specific vectors through directed evolution. Capsid genomes of adeno-associated virus (AAV) serotypes 1-9 were randomly fragmented and reassembled using PCR to generate a chimeric capsid library. A single infectious clone (chimeric-1829) containing genome fragments from AAV1, 2, 8, and 9 was isolated from an integrin minus hamster melanoma cell line previously shown to have low permissiveness to AAV. Molecular modeling studies suggest that AAV2 contributes to surface loops at the icosahedral threefold axis of symmetry, while AAV1 and 9 contribute to two- and fivefold symmetry interactions, respectively. The C-terminal domain (AAV9) was identified as a critical structural determinant of melanoma tropism through rational mutagenesis. Chimeric-1829 utilizes heparan sulfate as a primary receptor and transduces melanoma cells more efficiently than all serotypes. Further, chimeric-1829 demonstrates altered tropism in rodent skeletal muscle, liver, and brain including nonhuman primates. We determined a unique immunological profile based on neutralizing antibody (NAb) titer and crossreactivity studies strongly supporting isolation of a synthetic laboratory-derived capsid variant. Application of this technology to alternative cell/tissue types using AAV or other viral capsid sequences is likely to yield a new class of biological nanoparticles as vectors for human gene transfer.


Assuntos
Embaralhamento de DNA , Dependovirus/genética , Vetores Genéticos/isolamento & purificação , Genoma Viral/genética , Nanopartículas , Animais , Anticorpos/imunologia , Encéfalo/metabolismo , Capsídeo/imunologia , Cricetinae , Dependovirus/ultraestrutura , Evolução Molecular Direcionada , Biblioteca Gênica , Vetores Genéticos/genética , Humanos , Fígado/metabolismo , Melanoma , Camundongos , Camundongos Endogâmicos BALB C , Músculo Esquelético/metabolismo , Primatas , Transdução Genética , Internalização do Vírus
20.
Mol Ther ; 16(7): 1252-1260, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28178482

RESUMO

We report a DNA shuffling-based approach for developing cell type-specific vectors through directed evolution. Capsid genomes of adeno-associated virus (AAV) serotypes 1-9 were randomly fragmented and reassembled using PCR to generate a chimeric capsid library. A single infectious clone (chimeric-1829) containing genome fragments from AAV1, 2, 8, and 9 was isolated from an integrin minus hamster melanoma cell line previously shown to have low permissiveness to AAV. Molecular modeling studies suggest that AAV2 contributes to surface loops at the icosahedral threefold axis of symmetry, while AAV1 and 9 contribute to two- and fivefold symmetry interactions, respectively. The C-terminal domain (AAV9) was identified as a critical structural determinant of melanoma tropism through rational mutagenesis. Chimeric-1829 utilizes heparan sulfate as a primary receptor and transduces melanoma cells more efficiently than all serotypes. Further, chimeric-1829 demonstrates altered tropism in rodent skeletal muscle, liver, and brain including nonhuman primates. We determined a unique immunological profile based on neutralizing antibody (NAb) titer and crossreactivity studies strongly supporting isolation of a synthetic laboratory-derived capsid variant. Application of this technology to alternative cell/tissue types using AAV or other viral capsid sequences is likely to yield a new class of biological nanoparticles as vectors for human gene transfer.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...