Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Mov Disord ; 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38718138

RESUMO

OBJECTIVE: Gene therapy by convection-enhanced delivery of type 2 adeno-associated virus-glial cell derived neurotrophic factor (AAV2-GDNF) to the bilateral putamina seeks to increase GDNF gene expression and treat Parkinson's disease (PD). METHODS: A 63-year-old man with advanced PD received AAV2-GDNF in a clinical trial. He died from pneumonia after anterior cervical discectomy and fusion 45 months later. An autopsy included brain examination for GDNF transgene expression. Putaminal catecholamine concentrations were compared to in vivo 18F-Fluorodopa (18F-FDOPA) positron emission tomography (PET) scanning results before and 18 months after AAV2-GDNF infusion. RESULTS: Parkinsonian progression stabilized clinically. Postmortem neuropathology confirmed PD. Bilateral putaminal regions previously infused with AAV2-GDNF expressed the GDNF gene. Total putaminal dopamine was 1% of control, confirming the striatal dopaminergic deficiency suggested by baseline 18F-DOPA-PET scanning. Putaminal regions responded as expected to AAV2-GDNF. CONCLUSION: After AAV2-GDNF infusion, infused putaminal regions showed increased GDNF gene expression, tyrosine hydroxylase immunoreactive sprouting, catechol levels, and 18F-FDOPA-PET signal, suggesting the regenerative potential of AAV2-GDNF in PD. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.

2.
Mol Ther ; 26(10): 2418-2430, 2018 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-30057240

RESUMO

The present study was designed to characterize transduction of non-human primate brain and spinal cord with a modified adeno-associated virus serotype 2, incapable of binding to the heparan sulfate proteoglycan receptor, referred to as AAV2-HBKO. AAV2-HBKO was infused into the thalamus, intracerebroventricularly or via a combination of both intracerebroventricular and thalamic delivery. Thalamic injection of this modified vector encoding GFP resulted in widespread CNS transduction that included neurons in deep cortical layers, deep cerebellar nuclei, several subcortical regions, and motor neuron transduction in the spinal cord indicative of robust bidirectional axonal transport. Intracerebroventricular delivery similarly resulted in widespread cortical transduction, with one striking distinction that oligodendrocytes within superficial layers of the cortex were the primary cell type transduced. Robust motor neuron transduction was also observed in all levels of the spinal cord. The combination of thalamic and intracerebroventricular delivery resulted in transduction of oligodendrocytes in superficial cortical layers and neurons in deeper cortical layers. Several subcortical regions were also transduced. Our data demonstrate that AAV2-HBKO is a powerful vector for the potential treatment of a wide number of neurological disorders, and highlight that delivery route can significantly impact cellular tropism and pattern of CNS transduction.


Assuntos
Terapia Genética , Vetores Genéticos/efeitos adversos , Neurônios/efeitos dos fármacos , Parvovirinae/genética , Medula Espinal/efeitos dos fármacos , Animais , Transporte Axonal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Proteínas do Capsídeo/administração & dosagem , Proteínas do Capsídeo/genética , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/patologia , Dependovirus , Modelos Animais de Doenças , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Proteoglicanas de Heparan Sulfato/administração & dosagem , Proteoglicanas de Heparan Sulfato/genética , Humanos , Infusões Intraventriculares , Neurônios Motores/efeitos dos fármacos , Neurônios/patologia , Primatas , Medula Espinal/patologia , Tálamo/efeitos dos fármacos
3.
Mol Ther ; 21(1): 158-66, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22929660

RESUMO

There is considerable interest in the use of adeno-associated virus serotype 9 (AAV9) for neurological gene therapy partly because of its ability to cross the blood-brain barrier to transduce astrocytes and neurons. This raises the possibility that AAV9 might also transduce antigen-presenting cells (APC) in the brain and provoke an adaptive immune response. We tested this hypothesis by infusing AAV9 vectors encoding foreign antigens, namely human aromatic L-amino acid decarboxylase (hAADC) and green fluorescent protein (GFP), into rat brain parenchyma. Over ensuing weeks, both vectors elicited a prominent inflammation in transduced brain regions associated with upregulation of MHC II in glia and associated lymphocytic infiltration. Transduction of either thalamus or striatum with AAV9-hAADC evinced a significant loss of neurons and induction of anti-hAADC antibodies. We conclude that AAV9 transduces APC in the brain and, depending on the immunogenicity of the transgene, can provoke a full immune response that mediates significant brain pathology. We emphasize, however, that these observations do not preclude the use of AAV serotypes that can transduce APC. However, it does potentially complicate preclinical toxicology studies in which non-self proteins are expressed at a level sufficient to trigger cell-mediated and humoral immune responses.


Assuntos
Dependovirus/genética , Vetores Genéticos , Imunidade Celular , Proteínas/genética , Animais , Proteínas/imunologia , Ratos , Transdução Genética
4.
Nat Med ; 29(8): 2030-2040, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37580533

RESUMO

Alcohol use disorder (AUD) exacts enormous personal, social and economic costs globally. Return to alcohol use in treatment-seeking patients with AUD is common, engendered by a cycle of repeated abstinence-relapse episodes even with use of currently available pharmacotherapies. Repeated ethanol use induces dopaminergic signaling neuroadaptations in ventral tegmental area (VTA) neurons of the mesolimbic reward pathway, and sustained dysfunction of reward circuitry is associated with return to drinking behavior. We tested this hypothesis by infusing adeno-associated virus serotype 2 vector encoding human glial-derived neurotrophic factor (AAV2-hGDNF), a growth factor that enhances dopaminergic neuron function, into the VTA of four male rhesus monkeys, with another four receiving vehicle, following induction of chronic alcohol drinking. GDNF expression ablated the return to alcohol drinking behavior over a 12-month period of repeated abstinence-alcohol reintroduction challenges. This behavioral change was accompanied by neurophysiological modulations to dopamine signaling in the nucleus accumbens that countered the hypodopaminergic signaling state associated with chronic alcohol use, indicative of a therapeutic modulation of limbic circuits countering the effects of alcohol. These preclinical findings suggest gene therapy targeting relapse prevention may be a potential therapeutic strategy for AUD.


Assuntos
Alcoolismo , Animais , Masculino , Consumo de Bebidas Alcoólicas/genética , Consumo de Bebidas Alcoólicas/metabolismo , Alcoolismo/terapia , Alcoolismo/tratamento farmacológico , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Etanol/metabolismo , Etanol/farmacologia , Etanol/uso terapêutico , Terapia Genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Núcleo Accumbens/metabolismo , Primatas/genética , Área Tegmentar Ventral/metabolismo
5.
J Neurooncol ; 108(1): 53-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22350373

RESUMO

The mammalian target of rapamycin (mTOR) plays a central role in regulating the proliferation of cancer cells, and mTOR-specific inhibitors such as rapamycin analogs are considered as a promising therapy for malignant glioma. In this study, we investigated the possibility of using mTOR inhibitors to treat gliomas. We used a molecular marker, phosphorylation of S6 protein, to monitor biological effects of mTOR inhibitors within xenografts. Phosphorylation was decreased more in U87MG glioma after treatment with high doses of rapamycin or its analog, torisel (10 mg/kg or 25 mg/kg), but only slightly after a low dose of rapamycin (3 mg/kg). This effect correlated with enhanced survival of rats after weekly peritoneal injections of both drugs at the highest two doses but not at the low dose. High doses of both drugs caused weight loss in rats. Clinical trial data indicates that low doses of Torisel (<3 mg/kg) were not efficacious in recurrent GBM. It is concluded that systemic administration of rapamycin analogues may not be a treatment option for patients with malignant glioma due to the intolerability of high doses that might otherwise be effective. The present study underscores the need for better pre-clinical evaluation of drugs with respect to therapeutic window.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Glioma/tratamento farmacológico , Imunossupressores/administração & dosagem , Sirolimo/administração & dosagem , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Transplante de Neoplasias , Fosforilação/efeitos dos fármacos , Proteínas Quinases/metabolismo , Ratos , Ratos Nus , Fatores de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Mol Ther ; 19(5): 922-7, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21102559

RESUMO

We elucidated the effects of parkinsonian degeneration on trafficking of AAV2-GDNF in the nigro-striatum (nigro-ST) of unilaterally 6-hydroxydopamine (6-OHDA)-lesioned rats. Vector infused into striatum (ST) was transported to substantia nigra (SN), both pars compacta (SNc), and pars reticulata (SNr). In the lesioned hemisphere, glial cell line-derived neurotrophic factor (GDNF) immunoreactivity was only found in SNr consistent with elimination of SNc dopaminergic (DA) neurons by 6-OHDA. Further analysis showed that striatal delivery of AAV2-GDNF resulted in GDNF expression in globus pallidus (GP), entopeduncular nucleus (EPN), and subthalamic nucleus (STN) in both lesioned and unlesioned hemispheres. Injection of vector into SN, covering both SNc and SNr, resulted in striatal expression of GDNF in the unlesioned hemisphere but not in the lesioned hemisphere. No expression was seen in GP or EPN. We conclude that adeno-associated virus serotype 2 (AAV2) is transported throughout the nigro-ST exclusively by anterograde transport. This transport phenomenon directs GDNF expression throughout the basal ganglia in regions that are adversely affected in Parkinson's disease (PD) in addition to SNc. Delivery of vector to SN, however, does not direct expression of GDNF in ST, EPN, or GP. On this basis, we believe that striatal delivery of AAV2-GDNF is the preferred course of action for trophic rescue of DA function.


Assuntos
Transporte Axonal , Gânglios da Base/metabolismo , Dependovirus/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Animais , Transporte Axonal/efeitos dos fármacos , Núcleo Entopeduncular/citologia , Expressão Gênica , Terapia Genética/métodos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Globo Pálido/citologia , Masculino , Fatores de Crescimento Neural/metabolismo , Oxidopamina/farmacologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Doença de Parkinson/terapia , Ratos , Ratos Sprague-Dawley , Substância Negra/metabolismo , Substância Negra/patologia , Núcleo Subtalâmico/citologia
7.
Proc Natl Acad Sci U S A ; 106(7): 2407-11, 2009 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-19193857

RESUMO

Transduction of the primate cortex with adeno-associated virus (AAV)-based gene therapy vectors has been challenging, because of the large size of the cortex. We report that a single infusion of AAV2 vector into thalamus results in widespread expression of transgene in the cortex through transduction of widely dispersed thalamocortical projections. This finding has important implications for the treatment of certain genetic and neurodegenerative diseases.


Assuntos
Córtex Cerebral/metabolismo , Dependovirus/metabolismo , Terapia Genética/métodos , Doenças Neurodegenerativas/terapia , Animais , Vetores Genéticos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Humanos , Imuno-Histoquímica/métodos , Macaca mulatta , Modelos Biológicos , Modelos Genéticos , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Tálamo/metabolismo , Transgenes
8.
J Neurosci ; 30(28): 9567-77, 2010 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-20631185

RESUMO

Clinical studies to date have failed to establish therapeutic benefit of glial cell-derived neurotrophic factor (GDNF) in Parkinson's disease (PD). In contrast to previous nonclinical neuroprotective reports, this study shows clinically relevant and long-lasting regeneration of the dopaminergic system in rhesus macaques lesioned with 1-methy-4-phenyl-1,2,3,6-tetrahydropyridine 3-6 months before GDNF gene delivery (AAV2-GDNF). The observed progressive amelioration of functional deficits, recovery of dopamine, and regrowth of fibers to the striatal neuropil demonstrate that high GDNF expression in the putamen promotes restoration of the dopaminergic system in a primate model of advanced PD. Extensive distribution of GDNF within the putamen and transport to the severely lesioned substantia nigra, after convection-enhanced delivery of AAV2-GDNF into the putamen, indicates anterograde transport via striatonigral connections and is anticipated to occur in PD patients. Overall, these data demonstrate nonclinical neurorestoration after putaminal infusion of AAV2-GDNF and suggest that clinical investigation in PD patients is warranted.


Assuntos
Dopamina/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/uso terapêutico , Regeneração Nervosa/genética , Neurônios/metabolismo , Transtornos Parkinsonianos/terapia , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Análise de Variância , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Cromatografia Líquida de Alta Pressão , Ensaio de Imunoadsorção Enzimática , Feminino , Terapia Genética , Vetores Genéticos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Imuno-Histoquímica , Macaca mulatta , Masculino , Neurônios/patologia , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/patologia , Recuperação de Função Fisiológica
9.
Neuroimage ; 54 Suppl 1: S189-95, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20080195

RESUMO

Recently, we developed an MRI-based method that enables tracking of parenchymal infusions of therapeutic agents by inclusion of a contrast reagent in the infusate. We show that both liposomal Gadoteridol (GDL) and free Gadoteridol (Gd) can be used for MRI-monitored infusions into the non-human primate (NHP) putamen to predict the distribution of GDNF protein after convection-enhanced delivery (CED). GDNF and both MRI tracers showed good co-distribution within the putamen and other brain regions. Although the CED infusion technique can distribute GDNF protein over large brain regions, continuous administration of GDNF could cause undesired effects that could counteract the benefits of CED as demonstrated in this study when large volumes of GDNF were delivered that lead to GDNF leakage into CSF. These limitations can be addressed by employing an intermittent CED schedule that permits consistent target coverage without GDNF leakage into CSF or white matter. We present an approach intracranial GDNF infusions that can be optimized by means of real-time monitoring via MRI. Adoption of this new standard, along with advanced, reflux-resistant cannulae, may permit reconsideration of direct GDNF infusion into parenchyma as a clinical strategy, since previous clinical studies involving chronic infusion of recombinant glial cell line-derived neurotrophic factor (GDNF) to the putamen for the treatment of Parkinson's disease have yielded mixed results, a state of affairs that may in part be attributed to suboptimal infusion parameters.


Assuntos
Meios de Contraste/administração & dosagem , Fator Neurotrófico Derivado de Linhagem de Célula Glial/administração & dosagem , Compostos Heterocíclicos/administração & dosagem , Compostos Organometálicos/administração & dosagem , Putamen/metabolismo , Cirurgia Assistida por Computador/métodos , Animais , Meios de Contraste/farmacocinética , Convecção , Sistemas de Liberação de Medicamentos/métodos , Gadolínio , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacocinética , Compostos Heterocíclicos/farmacocinética , Macaca fascicularis , Imageamento por Ressonância Magnética , Masculino , Compostos Organometálicos/farmacocinética , Distribuição Tecidual
10.
Mol Ther ; 18(8): 1458-61, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20531394

RESUMO

This study completes the longest known in vivo monitoring of adeno-associated virus (AAV)-mediated gene expression in nonhuman primate (NHP) brain. Although six of the eight parkinsonian NHP originally on study have undergone postmortem analysis, as described previously, we monitored the remaining two animals for a total of 8 years. In this study, NHP received AAV2-human L-amino acid decarboxylase (hAADC) infusions into the MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-lesioned putamen. Restoration of AADC activity restored normal response to levodopa and gene expression could be quantitated repeatedly over many years by 6-[(18)F]fluoro-meta-tyrosine (FMT)-positron emission tomography (PET) and confirm that AADC transgene expression remained unchanged at the 8-year point. Behavioral assessments confirmed continued, normalized response to levodopa (improvement by 35% over historical controls). Postmortem analysis showed that, although only 5.6 + or - 1% and 6.6 + or - 1% of neurons within the transduced volumes of the striatum were transduced, this still secured robust clinical improvement. Importantly, there were no signs of neuroinflammation or reactive gliosis at the 8-year point, indicative of the safety of this treatment. The present data suggest that the improvement in the L-3,4-dihydroxyphenylalanine (L-Dopa) therapeutic window brought about by AADC gene therapy is pronounced and persistent for many years.


Assuntos
Descarboxilases de Aminoácido-L-Aromático/metabolismo , Dependovirus/genética , Macaca mulatta/metabolismo , Animais , Descarboxilases de Aminoácido-L-Aromático/genética , Encéfalo/metabolismo , Corpo Estriado/metabolismo , Dopamina/metabolismo , Imuno-Histoquímica , Levodopa/metabolismo , Masculino , Microscopia de Fluorescência
11.
Mol Ther ; 18(8): 1490-5, 2010 08.
Artigo em Inglês | MEDLINE | ID: mdl-20551915

RESUMO

Gene therapies that utilize convention-enhanced delivery (CED) will require close monitoring of vector infusion in real time and accurate prediction of drug distribution. The magnetic resonance imaging (MRI) contrast agent, Gadoteridol (Gd), was used to monitor CED infusion and to predict the expression pattern of glial cell line-derived neurotrophic factor (GDNF) protein after administration of adeno-associated virus type 2 (AAV2) vector encoding human pre-pro-GDNF complementary DNA. The nonhuman primate (NHP) thalamus was utilized for modeling infusion to allow delivery of volumes more relevant to planned human studies. AAV2 encoding human aromatic L-amino acid decarboxylase (AADC) was coinfused with AAV2-GDNF/Gd to confirm regions of AAV2 transduction versus extracellular GDNF diffusion. There was a close correlation between Gd distribution and GDNF or AADC expression, and the ratios of expression areas of GDNF or AADC versus Gd were both close to 1. Our data support the use of Gd and MRI to monitor AAV2 infusion via CED and to predict the distribution of GDNF protein after AAV2-GDNF administration.


Assuntos
Adenoviridae/genética , Meios de Contraste , Compostos Heterocíclicos , Imageamento por Ressonância Magnética/métodos , Compostos Organometálicos , Animais , Descarboxilases de Aminoácido-L-Aromático/genética , Descarboxilases de Aminoácido-L-Aromático/metabolismo , Gadolínio , Vetores Genéticos/genética , Humanos , Macaca mulatta
12.
Chem Sci ; 11(33): 8973-8980, 2020 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-34123152

RESUMO

Reduction sensitive linkers (RSLs) have the potential to transform the field of drug delivery due to their ease of use and selective cleavage in intracellular environments. However, despite their compelling attributes, developing reduction sensitive self-immolative linkers for aliphatic amines has been challenging due to their poor leaving group ability and high pK a values. Here a traceless self-immolative linker composed of a dithiol-ethyl carbonate connected to a benzyl carbamate (DEC) is presented, which can modify aliphatic amines and release them rapidly and quantitatively after disulfide reduction. DEC was able to reversibly modify the lysine residues on CRISPR-Cas9 with either PEG, the cell penetrating peptide Arg10, or donor DNA, and generated Cas9 conjugates with significantly improved biological properties. In particular, Cas9-DEC-PEG was able to diffuse through brain tissue significantly better than unmodified Cas9, making it a more suitable candidate for genome editing in animals. Furthermore, conjugation of Arg10 to Cas9 with DEC was able to generate a self-delivering Cas9 RNP that could edit cells without transfection reagents. Finally, conjugation of donor DNA to Cas9 with DEC increased the homology directed DNA repair (HDR) rate of the Cas9 RNP by 50% in HEK 293T cell line. We anticipate that DEC will have numerous applications in biotechnology, given the ubiquitous presence of aliphatic amines on small molecule and protein therapeutics.

13.
Neuroimage ; 47 Suppl 2: T27-35, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19095069

RESUMO

We are developing a method for real-time magnetic resonance imaging (MRI) visualization of convection-enhanced delivery (CED) of adeno-associated viral vectors (AAV) to the primate brain. By including gadolinium-loaded liposomes (GDL) with AAV, we can track the convective movement of viral particles by continuous monitoring of distribution of surrogate GDL. In order to validate this approach, we infused two AAV (AAV1-GFP and AAV2-hAADC) into three different regions of non-human primate brain (corona radiata, putamen, and thalamus). The procedure was tolerated well by all three animals in the study. The distribution of GFP determined by immunohistochemistry in both brain regions correlated closely with distribution of GDL determined by MRI. Co-distribution was weaker with AAV2-hAADC, although in vivo PET scanning with FMT for AADC activity correlated well with immunohistochemistry of AADC. Although this is a relatively small study, it appears that AAV1 correlates better with MRI-monitored delivery than does AAV2. It seems likely that the difference in distribution may be due to differences in tissue specificity of the two serotypes.


Assuntos
Encéfalo/diagnóstico por imagem , Encéfalo/virologia , Dependovirus/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Animais , Descarboxilases de Aminoácido-L-Aromático/genética , Descarboxilases de Aminoácido-L-Aromático/metabolismo , Encéfalo/fisiologia , Gadolínio , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imuno-Histoquímica , Lipossomos , Macaca mulatta , Imageamento por Ressonância Magnética , Masculino , Tomografia por Emissão de Pósitrons , Putamen/virologia , Tálamo/virologia
14.
J Neurooncol ; 95(2): 185-197, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19466380

RESUMO

Convection-enhanced delivery (CED) of highly stable PEGylated liposomes encapsulating chemotherapeutic drugs has previously been effective against malignant glioma xenografts. We have developed a novel, convectable non-PEGylated liposomal formulation that can be used to encapsulate both the topoisomerase I inhibitor topotecan (topoCED) and paramagnetic gadodiamide (gadoCED), providing an ideal basis for real-time monitoring of drug distribution. Tissue retention of topoCED following single CED administration was significantly improved relative to free topotecan. At a dose of 10 microg (0.5 mg/ml), topoCED had a half-life in brain of approximately 1 day and increased the area under the concentration-time curve (AUC) by 28-fold over free topotecan (153.8 vs. 5.5 microg day/g). The combination of topoCED and gadoCED was found to co-convect well in both naïve rat brain and malignant glioma xenografts (correlation coefficients 0.97-0.99). In a U87MG cell assay, the 50% inhibitory concentration (IC(50)) of topoCED was approximately 0.8 microM at 48 and 72 h; its concentration-time curves were similar to free topotecan and unaffected by gadoCED. In a U87MG intracranial rat xenograft model, a two-dose CED regimen of topoCED co-infused with gadoCED greatly increased median overall survival at dose levels of 0.5 mg/ml (29.5 days) and 1.0 mg/ml (33.0 days) vs. control (20.0 days; P < 0.0001 for both comparisons). TopoCED at higher concentrations (1.6 mg/ml) co-infused with gadoCED showed no evidence of histopathological changes attributable to either agent. The positive results of tissue pharmacokinetics, co-convection, cytotoxicity, efficacy, and lack of toxicity of topoCED in a clinically meaningful dose range, combined with an ideal matched-liposome paramagnetic agent, gadoCED, implicates further clinical applications of this therapy in the treatment of malignant glioma.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Gadolínio DTPA/administração & dosagem , Glioblastoma/tratamento farmacológico , Polietilenoglicóis/química , Topotecan/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Neoplasias Encefálicas/patologia , Sobrevivência Celular/efeitos dos fármacos , Meios de Contraste/administração & dosagem , Meios de Contraste/farmacocinética , Convecção , Gadolínio DTPA/farmacocinética , Glioblastoma/patologia , Humanos , Lipossomos , Masculino , Ratos , Ratos Sprague-Dawley , Taxa de Sobrevida , Distribuição Tecidual , Topotecan/farmacocinética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Mol Ther ; 16(8): 1392-9, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18545225

RESUMO

In Parkinson's disease (PD) chronic inflammation occurs in the substantia nigra (SNc) concurrently with dopaminergic neurodegeneration. In models of PD, microglial activation precedes neurodegeneration in the SNc, suggesting that the underlying pathogenesis involves a complex response in the nigrostriatal pathway, and that the innate immune system plays a significant role. We have investigated the neuroprotective effect of an adeno-associated viral type-2 (AAV2) vector containing the complementary DNA (cDNA) for human interleukin-10 (hIL-10) in the unilateral 6-hydroxydopamine (6-OHDA) rat model of PD. AAV2-hIL-10 reduced the 6-OHDA-induced loss of tyrosine hydroxylase (TH)-positive neurons in the SNc, and also reduced loss of striatal dopamine (DA). Pretreatment with AAV2-hIL-10 reduced glial activation in the SNc but did not attenuate striatal release of the inflammatory cytokine IL-1beta. Assessment of rotational behavior in response to apomorphine challenge showed absence of asymmetry, confirming protection of dopaminergic innervation of the lesioned striatum. At baseline, 6-OHDA-lesioned animals displayed a deficit in contralateral forelimb use, but pretreatment with AAV2-hIL-10 reduced this forelimb akinesia. Transcriptional analyses revealed alteration of a few genes by AAV2-hIL-10; these alterations may contribute to neuroprotection. This study supports the need for further investigations relating to gene therapies aimed at reducing neuroinflammation in early PD.


Assuntos
Modelos Animais de Doenças , Terapia Genética/métodos , Interleucina-10/fisiologia , Doença de Parkinson/terapia , Animais , Apomorfina/farmacologia , Comportamento Animal/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Dependovirus/genética , Ensaio de Imunoadsorção Enzimática , Membro Anterior/efeitos dos fármacos , Membro Anterior/metabolismo , Membro Anterior/fisiopatologia , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Vetores Genéticos/genética , Humanos , Hidroxidopaminas/farmacologia , Imuno-Histoquímica , Interleucina-10/genética , Interleucina-10/metabolismo , Masculino , Doença de Parkinson/genética , Doença de Parkinson/fisiopatologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo , Substância Negra/patologia
16.
Sci Transl Med ; 11(506)2019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31434754

RESUMO

Niemann-Pick disease type A (NPD-A) is a lysosomal storage disorder characterized by neurodegeneration and early death. It is caused by loss-of-function mutations in the gene encoding for acid sphingomyelinase (ASM), which hydrolyzes sphingomyelin into ceramide. Here, we evaluated the safety of cerebellomedullary (CM) cistern injection of adeno-associated viral vector serotype 9 encoding human ASM (AAV9-hASM) in nonhuman primates (NHP). We also evaluated its therapeutic benefit in a mouse model of the disease (ASM-KO mice). We found that CM injection in NHP resulted in widespread transgene expression within brain and spinal cord cells without signs of toxicity. CM injection in the ASM-KO mouse model resulted in hASM expression in cerebrospinal fluid and in different brain areas without triggering an inflammatory response. In contrast, direct cerebellar injection of AAV9-hASM triggered immune response. We also identified a minimally effective therapeutic dose for CM injection of AAV9-hASM in mice. Two months after administration, the treatment prevented motor and memory impairment, sphingomyelin (SM) accumulation, lysosomal enlargement, and neuronal death in ASM-KO mice. ASM activity was also detected in plasma from AAV9-hASM CM-injected ASM-KO mice, along with reduced SM amount and decreased inflammation in the liver. Our results support CM injection for future AAV9-based clinical trials in NPD-A as well as other lysosomal storage brain disorders.


Assuntos
Dependovirus/metabolismo , Terapia Genética , Doença de Niemann-Pick Tipo A/genética , Doença de Niemann-Pick Tipo A/terapia , Sorogrupo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Inflamação/patologia , Injeções , Fígado/patologia , Camundongos Knockout , Atividade Motora , Primatas , Esfingomielina Fosfodiesterase/administração & dosagem , Esfingomielina Fosfodiesterase/sangue , Esfingomielina Fosfodiesterase/genética , Transgenes
17.
Mol Ther Methods Clin Dev ; 13: 47-54, 2019 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-30666308

RESUMO

Here we evaluated the utility of MRI to monitor intrathecal infusions in nonhuman primates. Adeno-associated virus (AAV) spiked with gadoteridol, a gadolinium-based MRI contrast agent, enabled real-time visualization of infusions delivered either via cerebromedullary cistern, lumbar, cerebromedullary and lumbar, or intracerebroventricular infusion. The kinetics of vector clearance from the cerebrospinal fluid (CSF) were analyzed. Our results highlight the value of MRI in optimizing the delivery of infusate into CSF. In particular, MRI revealed differential patterns of infusate distribution depending on the route of delivery. Gadoteridol coverage analysis showed that cerebellomedullary cistern delivery was a reliable and effective route of injection, achieving broad infusate distribution in the brain and spinal cord, and was even greater when combined with lumbar injection. In contrast, intracerebroventricular injection resulted in strong cortical coverage but little spinal distribution. Lumbar injection alone led to the distribution of MRI contrast agent mainly in the spinal cord with little cortical coverage, but this delivery route was unreliable. Similarly, vector clearance analysis showed differences between different routes of delivery. Overall, our data support the value of monitoring CSF injections to dissect different patterns of gadoteridol distribution based on the route of intrathecal administration.

18.
Front Neurosci ; 12: 45, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29459819

RESUMO

Here we advance the hypothesis that Parkinson's disease (PD) is fundamentally a failure of trophic support for specific classes of neurons, primarily catecholaminergic. Evidence from our laboratory provides a framework into which a broad array of findings from many quarters can be integrated into a general theory that offers testable hypotheses to new and established investigators. Mice deficient in the ability to synthesize series-a gangliosides, specifically GM1 ganglioside, develop parkinsonism. We found that this seems to be due to a failure in signaling efficiency by the important catecholaminergic growth factor, GDNF. Interestingly, these mice accumulate alpha-synuclein in nigral neurons. Striatal over-expression of GDNF eliminates these aggregates and also restores normal motor function. These findings bring into question common beliefs about alpha-synuclein pathology and may help us to reinterpret other experimental findings in a new light. The purpose of this article is to provoke new thinking about PD and hopefully encourage younger scientists to explore some of the ideas presented below.

19.
Hum Gene Ther ; 17(3): 291-302, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16544978

RESUMO

Adeno-associated virus type 2 (AAV2)-based vectors are promising transgene carriers for experimental gene therapy treatments of brain diseases. However, detailed evaluation of transgene distribution, trafficking, and transport within the brain is of the utmost importance before applying any type of gene therapy in humans. We examined the distribution of AAV2-thymidine kinase (AAV2-TK) and AAV2-aromatic L-amino acid decarboxylase (AAV2-AADC) in monkey brain after convection-enhanced delivery (CED). The AADC group consisted of two 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned monkeys that received unilateral infusions of AAV2-AADC into six sites in the right hemisphere. The TK group consisted of three monkeys that received bilateral CED infusion of AAV2-TK into the putamen; one side in all three monkeys was coinfused with heparin. Six weeks after AAV delivery, the brains were collected and processed for immunohistochemical staining. Volumetric measurement of TK distribution showed that at least 75% of the putamen could be covered by a single infusion of the vector; however, no effects of heparin coadministration were found, most likely because of the already robust gene transfer achieved by CED. Interestingly, TK- and AADCimmunoreactive cells were also present outside the striatum, in the globus pallidus, subthalamic nucleus, thalamus, and substantia nigra. CED proved to be an efficient method for delivery of the AAV2 vector. Detection of the transgenes in brain structures distant from the site of injection emphasizes the potential for gene transport, and the advantages and disadvantages of CED for gene therapy deserve further study.


Assuntos
Transporte Biológico , Convecção , Dependovirus/genética , Putamen/enzimologia , Timidina Quinase/genética , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/administração & dosagem , Animais , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos , Heparina/administração & dosagem , Humanos , Macaca mulatta , Masculino , Doença de Parkinson/etiologia , Doença de Parkinson/patologia , Doença de Parkinson/terapia , Putamen/patologia , Timidina Quinase/metabolismo , Transfecção , Transgenes/fisiologia
20.
J Chem Neuroanat ; 78: 25-33, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27515691

RESUMO

Focus on the purinergic receptor P2Y11 has increased following the finding of an association between the sleep disorder narcolepsy and a genetic variant in P2RY11 causing decreased gene expression. Narcolepsy is believed to arise from an autoimmune destruction of the hypothalamic neurons that produce the neuropeptide hypocretin/orexin. It is unknown how a decrease in expression of P2Y11 might contribute to an autoimmune reaction towards the hypocretin neurons and the development of narcolepsy. To advance narcolepsy research it is therefore extremely important to determine the neuroanatomical localization of P2Y11 in the brain with particular emphasis on the hypocretin neurons. In this article we used western blot, staining of blood smears, and flow cytometry to select two antibodies for immunohistochemical staining of macaque monkey brain. Staining was seen in neuron-like structures in cortical and hypothalamic regions. Rats do not have a gene orthologue to the P2Y11 receptor and therefore rat brain was used as negative control tissue. The chromogenic signal observed in macaque monkey brain in neurons was not considered reliable, because the antibodies stained rat brain in a similar distribution pattern. Hence, the neuroanatomical localization of the P2Y11 receptor remains undetermined due to the lack of specific P2Y11 antibodies for brain immunohistochemistry.


Assuntos
Cerebelo/metabolismo , Hipotálamo/metabolismo , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Receptores Purinérgicos P2/metabolismo , Animais , Imuno-Histoquímica/métodos , Macaca , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA