Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
1.
NMR Biomed ; 26(1): 43-50, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22711560

RESUMO

The detection of Parkinson's disease (PD) in its preclinical stages prior to outright neurodegeneration is essential to the development of neuroprotective therapies and could reduce the number of misdiagnosed patients. However, early diagnosis is currently hampered by lack of reliable biomarkers. (1) H magnetic resonance spectroscopy (MRS) offers a noninvasive measure of brain metabolite levels that allows the identification of such potential biomarkers. This study aimed at using MRS on an ultrahigh field 14.1 T magnet to explore the striatal metabolic changes occurring in two different rat models of the disease. Rats lesioned by the injection of 6-hydroxydopamine (6-OHDA) in the medial-forebrain bundle were used to model a complete nigrostriatal lesion while a genetic model based on the nigral injection of an adeno-associated viral (AAV) vector coding for the human α-synuclein was used to model a progressive neurodegeneration and dopaminergic neuron dysfunction, thereby replicating conditions closer to early pathological stages of PD. MRS measurements in the striatum of the 6-OHDA rats revealed significant decreases in glutamate and N-acetyl-aspartate levels and a significant increase in GABA level in the ipsilateral hemisphere compared with the contralateral one, while the αSyn overexpressing rats showed a significant increase in the GABA striatal level only. Therefore, we conclude that MRS measurements of striatal GABA levels could allow for the detection of early nigrostriatal defects prior to outright neurodegeneration and, as such, offers great potential as a sensitive biomarker of presymptomatic PD.


Assuntos
Encéfalo/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Oxidopamina , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/metabolismo , alfa-Sinucleína , Ácido gama-Aminobutírico/análise , Animais , Biomarcadores/análise , Diagnóstico Precoce , Feminino , Transtornos Parkinsonianos/diagnóstico , Prótons , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
2.
Neuroimage ; 61(4): 1336-46, 2012 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-22450300

RESUMO

One of the core pathological features of Alzheimer's disease (AD) is the accumulation of amyloid plaques in the brain. Current efforts of medical imaging research aim at visualizing amyloid plaques in living patients in order to evaluate the progression of the pathology, but also to facilitate the diagnosis of AD at the prodromal stage. In this study, we evaluated the capabilities of a new experimental imaging setup to image amyloid plaques in the brain of a transgenic mouse model of Alzheimer's disease. This imaging setup relies on a grating interferometer at a synchrotron X-ray source to measure the differential phase contrast between brain tissue and amyloid plaques. It provides high-resolution images with a large field of view, making it possible to scan an entire mouse brain. Here, we showed that this setup yields sufficient contrast to detect amyloid plaques and to quantify automatically several important structural parameters, such as their size and their regional density in 3D, on the scale of a whole mouse brain. Whilst future developments are required to apply this technique in vivo, this grating-based setup already gives the possibility to perform powerful studies aiming at quantifying the amyloid pathology in mouse models of AD and might accelerate the evaluation of anti-amyloid compounds. In addition, this technique may also facilitate the development of other amyloid imaging methods such as positron emission tomography (PET) by providing convenient high-resolution 3D data of the plaque distribution for multimodal comparison.


Assuntos
Doença de Alzheimer/diagnóstico por imagem , Imageamento Tridimensional/métodos , Neuroimagem/métodos , Placa Amiloide/diagnóstico por imagem , Tomografia Computadorizada por Raios X/métodos , Doença de Alzheimer/patologia , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Modelos Animais de Doenças , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Transgênicos , Placa Amiloide/patologia
3.
Hum Mol Genet ; 21(8): 1861-76, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22246294

RESUMO

Mitochondrial dysfunction and oxidative stress have been implicated in the etiology of Parkinson's disease. Therefore, pathways controlling mitochondrial activity rapidly emerge as potential therapeutic targets. Here, we explore the neuronal response to prolonged overexpression of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α), a transcriptional regulator of mitochondrial function, both in vitro and in vivo. In neuronal primary cultures from the ventral midbrain, PGC-1α induces mitochondrial biogenesis and increases basal respiration. Over time, we observe an increasing proportion of the oxygen consumed by neurons which are dedicated to adenosine triphosphate production. In parallel to enhanced oxidative phosphorylation, PGC-1α progressively leads to a decrease in mitochondrial polarization. In the adult rat nigrostriatal system, adeno-associated virus (AAV)-mediated overexpression of PGC-1α induces the selective loss of dopaminergic markers and increases dopamine (DA) catabolism, leading to a reduction in striatal DA content. In addition, PGC-1α prevents the labeling of nigral neurons following striatal injection of the fluorogold retrograde tracer. When PGC-1α is expressed at higher levels following intranigral AAV injection, it leads to overt degeneration of dopaminergic neurons. Finally, PGC-1α overexpression does not prevent nigrostriatal degeneration in pathologic conditions induced by α-synuclein overexpression. Overall, we find that lasting overexpression of PGC-1α leads to major alterations in the metabolic activity of neuronal cells which dramatically impair dopaminergic function in vivo. These results highlight the central role of PGC-1α in the function and survival of dopaminergic neurons and the critical need for maintaining physiological levels of PGC-1α activity.


Assuntos
Neurônios Dopaminérgicos/fisiologia , Substância Negra/fisiologia , Transativadores/genética , Transativadores/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Respiração Celular , Sobrevivência Celular , Células Cultivadas , Corpo Estriado/metabolismo , Corpo Estriado/fisiologia , Dependovirus/genética , Dopamina/metabolismo , Neurônios Dopaminérgicos/citologia , Feminino , Regulação da Expressão Gênica , Vetores Genéticos , Humanos , Potencial da Membrana Mitocondrial , Camundongos , Mitocôndrias/metabolismo , Degeneração Neural , Neurônios/fisiologia , Fosforilação Oxidativa , Consumo de Oxigênio , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ratos , Ratos Sprague-Dawley , Substância Negra/citologia , Fatores de Transcrição , alfa-Sinucleína/metabolismo
4.
J Parkinsons Dis ; 1(4): 373-87, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-23939344

RESUMO

Although the overabundance of human alpha-synuclein in nigral dopaminergic neurons is considered to play a pathogenic role in Parkinson's disease (PD), it remains unclear how alpha-synuclein leads to neuronal degeneration and motor symptoms. Here, we explored the effect of human alpha-synuclein in the rat substantia nigra following AAV-mediated gene delivery inducing a moderate loss of dopaminergic neurons together with motor impairments. A significant fraction of the surviving nigral neurons were found to express human αSyn and displayed a pathological fragmentation of the Golgi apparatus. This observation prompted further investigation on the role of the secretory pathway, in particular at the ER/Golgi level, in alpha-synuclein toxicity. To address this question, we co-expressed human alpha-synuclein with Rab1A, a regulator of ER-to-Golgi vesicular trafficking, and found a significant reduction of Golgi fragmentation. Rab1A did not protect the dopaminergic neurons from the alpha-synuclein-induced degeneration that occurred within several months following vector injection. However, we observed in animals co-expressing Rab1A an improvement of motor behavior that correlates with the rescue of normal Golgi morphology in alpha-synuclein-expressing dopaminergic neurons. The non-prenylable mutant Rab1A-DeltaCC did not produce any of the effects observed with the wild-type form of Rab1A, linking the protective role of Rab1A with its activity in ER-to-Golgi vesicular trafficking. In conclusion, Rab1A can rescue the Golgi fragmentation caused by the overabundance of alpha-synuclein in nigral dopaminergic neurons, improving the ability of the surviving neurons to control motor function in hemiparkinsonian animals.


Assuntos
Neurônios Dopaminérgicos/ultraestrutura , Complexo de Golgi/patologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , alfa-Sinucleína/metabolismo , Proteínas rab1 de Ligação ao GTP/metabolismo , Animais , Apomorfina , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Comportamento Exploratório/efeitos dos fármacos , Feminino , Membro Anterior/fisiopatologia , Vetores Genéticos/fisiologia , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Humanos , Masculino , Camundongos , Mutação/genética , Doença de Parkinson/etiologia , Doença de Parkinson/fisiopatologia , Ratos , Ratos Sprague-Dawley , Substância Negra/efeitos dos fármacos , Substância Negra/patologia , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/genética
5.
Gene Ther ; 17(1): 141-6, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19727139

RESUMO

Retrograde transport of viral vectors in the rodent spinal cord provides a powerful means to administer a therapeutic transgene from the innervated musculature. With the aim of scaling up this approach to non-human primates, we have injected recombinant adeno-associated vectors (rAAV) serotype 6 expressing enhanced green fluorescent protein (eGFP) into the gastrocnemius muscle of African green monkeys to determine whether this results in efficient transgene delivery to lumbar motor neurons. Cells expressing eGFP were observed across more than 1 cm of the spinal cord 4 weeks after intramuscular injection, reaching more than half of motor neurons in some cross-sections. Furthermore, quantitative PCR on the spinal cord tissue confirmed that eGFP expression within motor neurons was due to bona fide retrograde transport of the vector genome from the muscle. Although infiltrations of macrophages and lymphocytes were observed in the rAAV2/6-injected muscle, there was no detectable immune response within the transduced region of the spinal cord. These findings imply that retrograde delivery of rAAV serotype 6 in a primate species constitutes a non-invasive and robust approach to transduce motor neurons, a crucial target cell population in neurodegenerative disorders, such as amyotrophic lateral sclerosis and spinal muscular atrophy.


Assuntos
Dependovirus/genética , Vetores Genéticos , Neurônios Motores/metabolismo , Transdução Genética , Animais , Chlorocebus aethiops , Proteínas de Fluorescência Verde/metabolismo , Injeções Intramusculares , Medula Espinal/citologia , Transgenes
6.
Neurobiol Dis ; 36(2): 303-11, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19660547

RESUMO

This study assessed the potential for functional and anatomical recovery of the diseased aged primate nigrostriatal system, in response to trophic factor gene transfer. Aged rhesus monkeys received a single intracarotid infusion of MPTP, followed one week later by MRI-guided stereotaxic intrastriatal and intranigral injections of lentiviral vectors encoding for glial derived neurotrophic factor (lenti-GDNF) or beta-galactosidase (lenti-LacZ). Functional analysis revealed that the lenti-GDNF, but not lenti-LacZ treated monkeys displayed behavioral improvements that were associated with increased fluorodopa uptake in the striatum ipsilateral to lenti-GDNF treatment. GDNF ELISA of striatal brain samples confirmed increased GDNF expression in lenti-GDNF treated aged animals that correlated with functional improvements and preserved nigrostriatal dopaminergic markers. Our results indicate that the aged primate brain challenged by MPTP administration has the potential to respond to trophic factor delivery and that the degree of neuroprotection depends on GDNF levels.


Assuntos
Envelhecimento , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Transtornos Parkinsonianos/genética , Fatores Etários , Envelhecimento/genética , Animais , Corpo Estriado/química , Corpo Estriado/metabolismo , Vetores Genéticos/administração & dosagem , Vetores Genéticos/uso terapêutico , Fator Neurotrófico Derivado de Linhagem de Célula Glial/administração & dosagem , Fator Neurotrófico Derivado de Linhagem de Célula Glial/biossíntese , Lentivirus/genética , Macaca mulatta , Masculino , Transtornos Parkinsonianos/fisiopatologia , Transtornos Parkinsonianos/prevenção & controle , Desempenho Psicomotor/efeitos dos fármacos , Desempenho Psicomotor/fisiologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/genética
7.
J Comp Neurol ; 515(1): 31-40, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19399891

RESUMO

In spite of partial success in treating Parkinson's disease by using ectopically placed grafts of dopamine-producing cells, restoration of the original neuroanatomical circuits, if possible, might work better. Previous evidence of normal anatomic projections from ventral mesencephalic (VM) grafts placed in the substantia nigra (SN) has been limited to neonatal rodents and double grafting or bridging procedures. This study attempted to determine whether injection of a potent growth-promoting factor, glial cell line-derived neurotrophic factor (GDNF), into the target regions or placement of fetal striatal co-grafts in the nigrostriatal pathway might elicit neuritic outgrowth to the caudate nucleus. Four adult St. Kitts green monkeys received embryonic VM grafts into the rostral mesencephalon near the host SN, and injections of adeno-associated virus 2 (AAV2)/GDNF or equine infectious anemia virus (EIAV)/GDNF into the caudate. Three adult monkeys were co-grafted with fetal VM tissue near the SN and fetal striatal grafts (STR) 2.5 mm rostral in the nigrostriatal pathway. Before sacrifice, the striatal target regions were injected with the retrograde tracer Fluoro-Gold (FG). FG label was found in tyrosine hydroxylase-labeled neurons in VM grafts in the SN of only those monkeys that received AAV2/GDNF vector injections into the ipsilateral striatum. All monkeys showed FG labeling in the host SN when FG labeling was injected on the same side. These data show that grafted dopaminergic neurons can extend neurites to a distant target releasing an elevated concentration of GDNF, and suggest that grafted neurons can be placed into appropriate loci for potential tract reconstruction.


Assuntos
Transplante de Tecido Encefálico/métodos , Corpo Estriado/metabolismo , Células-Tronco Embrionárias/transplante , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Transplante de Células-Tronco/métodos , Substância Negra/transplante , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Chlorocebus aethiops , Corpo Estriado/citologia , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Sobrevivência de Enxerto/fisiologia , Cones de Crescimento/metabolismo , Cones de Crescimento/ultraestrutura , Masculino , Vias Neurais/citologia , Vias Neurais/metabolismo , Neuritos/metabolismo , Neuritos/ultraestrutura , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/fisiopatologia , Transtornos Parkinsonianos/cirurgia , Coloração e Rotulagem , Estilbamidinas , Substância Negra/citologia , Substância Negra/metabolismo , Resultado do Tratamento , Regulação para Cima/fisiologia
8.
Hum Reprod ; 24(4): 790-6, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19273881

RESUMO

BACKGROUND: Animal studies have demonstrated better embryo development in vivo than in vitro. This pilot study tested the feasibility of using a novel in utero culture system (IUCS) to obtain normal human fertilization and embryo development. METHODS: The IUCS device comprised a perforated silicone hollow tube. The study included 13 patients (<36 years) undergoing a first intracytoplasmic sperm injection (ICSI) treatment and 167 metaphase II oocytes in three groups. In Group 1, 1-2 h after ICSI, sibling oocytes were assigned to IUCS or conventional in vitro culture. The device was retrieved on Day 1, and all zygotes were cultured in vitro till Day 5. In Group 2, fertilized oocytes were assigned on Day 1, embryos retrieved on Day 3 and all embryos cultured till Day 5. In Group 3, after Day 0 assignment, embryos were retrieved on Day 3 for blastomere biopsy and fluorescence in situ hybridization (FISH) and cultured until Day 5. The highest quality blastocysts were transferred on Day 5. RESULTS: Fertilization and embryo development were comparable in the in vitro and IUCS arms, with a tendency towards better embryo quality in the IUCS. FISH analysis in Group 3 revealed more normal embryos using the IUCS (P = 0.049). Three clinical pregnancies and live births were obtained: two from the IUCS arm and one from the in vitro arm. CONCLUSIONS: Our pilot study shows that this new IUCS appears to be feasible and safe, supporting normal fertilization, embryo development and normal chromosomal segregation. Furthermore, live births are possible after the transient presence of a silicone device in the uterus. Clinicaltrials.gov: NCT00480103.


Assuntos
Técnicas de Cultura Embrionária/métodos , Técnicas de Cultura Embrionária/instrumentação , Transferência Embrionária , Desenvolvimento Embrionário , Desenho de Equipamento , Feminino , Humanos , Hibridização in Situ Fluorescente , Recém-Nascido , Masculino , Projetos Piloto , Gravidez , Silicones , Injeções de Esperma Intracitoplásmicas , Fatores de Tempo
9.
Exp Neurol ; 215(1): 191-200, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19022249

RESUMO

Huntington's disease (HD) is a neurodegenerative disorder resulting from the expansion of a glutamine repeat (polyQ) in the N-terminus of the huntingtin (htt) protein. Expression of polyQ-containing proteins has been previously shown to induce various cellular stress responses. Among these, activation of the c-Jun N-terminal kinase (JNK) cascade has been observed in cellular models of HD. However, the implication of the JNK pathway has not previously been evaluated in the striatum of HD animal models. Here we report that the JNK pathway participates in HD pathology in a rat model of the disease. Increased phosphorylation of the JNK target c-Jun was observed as early as 4 weeks and persisted for 13 weeks after lentiviral-mediated expression of htt171-82Q. In order to assess the importance of this pathway in HD pathology, JNK inhibitors including dominant-negative mutants of upstream kinases (ASK1(K709R), MEKK1(D1369A)), a c-Jun mutant (Delta169c-Jun) and the active domain of the scaffold protein JIP-1/IBI (IBI-JBD) were tested for their ability to mitigate the effect of htt171-82Q. The overexpression of MEKK1(D1369A) and JIP-1/IBI reduced the polyQ-related loss of DARPP-32 expression, while the other inhibitors had no effect. In all cases, the formation of EM48-positive htt inclusions and P-c-Jun immunoreactivity were unaltered. These results suggest that JNK activation is involved in HD and that blockade of this pathway may be of benefit in counteracting HD-related neurotoxicity.


Assuntos
Doença de Huntington/enzimologia , Doença de Huntington/fisiopatologia , MAP Quinase Quinase 4/metabolismo , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Proteína de Ligação a CREB/metabolismo , Linhagem Celular Transformada , Modelos Animais de Doenças , Dopamina/metabolismo , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Feminino , Regulação da Expressão Gênica/fisiologia , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Lentivirus/fisiologia , Mutação/fisiologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Ratos , Ratos Wistar , Serina/metabolismo , Fatores de Tempo , Transfecção/métodos
10.
Proc Natl Acad Sci U S A ; 103(15): 6007-12, 2006 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-16581901

RESUMO

The reasons for the cellular specificity and slow progression of motoneuron diseases such as ALS are still poorly understood. We previously described a motoneuron-specific cell death pathway downstream of the Fas death receptor, in which synthesis of nitric oxide (NO) is an obligate step. Motoneurons from ALS model mice expressing mutant SOD1 showed increased susceptibility to exogenous NO as compared with controls. Here, we report a signaling mechanism whereby NO leads to death of mutant, but not control, motoneurons. Unexpectedly, exogenous NO triggers expression of Fas ligand (FasL) in cultured motoneurons. In mutant SOD1(G93A) and SOD1(G85R), but not in control motoneurons, this up-regulation results in activation of Fas, leading through Daxx to phosphorylation of p38 and further NO synthesis. This Fas/NO feedback amplification loop is required for motoneuron death in vitro. In vivo, mutant SOD1(G93A) and SOD1(G85R) mice show increased numbers of positive motoneurons and Daxx nuclear bodies weeks before disease onset. Moreover, FasL up-regulation is reduced in the presence of transgenic dominant-negative Daxx. We propose that chronic low-level activation of the Fas/NO feedback loop may underlie the motoneuron loss that characterizes familial ALS and may help to explain its slowly progressive nature.


Assuntos
Proteínas de Transporte/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Glicoproteínas de Membrana/fisiologia , Doença dos Neurônios Motores/fisiopatologia , Neurônios Motores/fisiologia , Proteínas Nucleares/fisiologia , Fatores de Necrose Tumoral/fisiologia , Receptor fas/fisiologia , Animais , Proteínas de Transporte/genética , Proteínas Correpressoras , Cruzamentos Genéticos , Proteína Ligante Fas , Retroalimentação , Feminino , Homozigoto , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Modelos Animais , Chaperonas Moleculares , Doença dos Neurônios Motores/genética , Doença dos Neurônios Motores/patologia , Óxido Nítrico/fisiologia , Proteínas Nucleares/genética , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único , Medula Espinal/patologia , Medula Espinal/fisiopatologia , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Fatores de Necrose Tumoral/genética , Receptor fas/genética
11.
Gene Ther ; 13(6): 487-95, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16319945

RESUMO

Experimental recapitulation of recessive human genetic neurodegenerative disease in rodents can be classically addressed through genetic disruption of the related gene. Although very informative, this specific gene targeting is restricted to mice and precludes a species scale-up towards non-human primates. Concomitantly, this requirement to silence a specific gene in a broad range of animal models is important in the design of therapeutic approaches to dominantly inherited neurodegenerative diseases. The emergence of RNA interference (RNAi), a highly specific mechanism of post-translational gene silencing, has opened a plethora of biological application ranging from reverse genetic analysis to therapeutic schemes. Recombinant viral vectors, by promoting a long-lasting delivery of genetic instructions in a broad range of cellular types of different species origins, represent potential platforms mandating silencing of specific gene products through RNAi. This review aims at providing an overview of the different viral systems engineered so far for efficient in vitro and in vivo delivery of RNAi instructions. Additionally, the potential of RNAi for functional analysis and therapy for polyglutamine disorders or amyotrophic lateral sclerosis is discussed.


Assuntos
Terapia Genética/métodos , Doenças Neurodegenerativas/terapia , Interferência de RNA , Animais , Engenharia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Humanos , Modelos Animais , Viroses/genética , Vírus/genética
12.
Gene Ther ; 13(5): 379-88, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16355116

RESUMO

Glial cell line-derived neurotrophic factor (GDNF) has been shown to increase the survival and functioning of dopamine neurons in a variety of animal models and some recent human trials. However, delivery of any protein to the brain remains a challenge due to the blood/brain barrier. Here we show that human neural progenitor cells (hNPC) can be genetically modified to release glycosylated GDNF in vitro under an inducible promoter system. hNPC-GDNF were transplanted into the striatum of rats 10 days following a partial lesion of the dopamine system. At 2 weeks following transplantation, the cells had migrated within the striatum and were releasing physiologically relevant levels of GDNF. This was sufficient to increase host dopamine neuron survival and fiber outgrowth. At 5 weeks following grafting there was a strong trend towards functional improvement in transplanted animals and at 8 weeks the cells had migrated to fill most of the striatum and continued to release GDNF with transport to the substantia nigra. These cells could also survive and release GDNF 3 months following transplantation into the aged monkey brain. No tumors were found in any animal. hNPC can be genetically modified, and thereby represent a safe and powerful option for delivering growth factors to specific targets within the central nervous system for diseases such as Parkinson's.


Assuntos
Encéfalo/metabolismo , Terapia Genética/métodos , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Neurônios/fisiologia , Transtornos Parkinsonianos/terapia , Transplante de Células-Tronco/métodos , Animais , Western Blotting/métodos , Dopamina/metabolismo , Vetores Genéticos/administração & dosagem , Fator Neurotrófico Derivado de Linhagem de Célula Glial/análise , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Haplorrinos , Humanos , Imuno-Histoquímica/métodos , Lentivirus/genética , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Células-Tronco/fisiologia , Transdução Genética/métodos
13.
Hum Gene Ther ; 15(10): 968-75, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15585112

RESUMO

Huntington's disease (HD) is a monogenic neurodegenerative disease that affects the efferent neurons of the striatum. The protracted evolution of the pathology over 15 to 20 years, after clinical onset in adulthood, underscores the potential of therapeutic tools that would aim at protecting striatal neurons. Proteins with neuroprotective effects in the adult brain have been identified, among them ciliary neurotrophic factor (CNTF), which protected striatal neurons in animal models of HD. Accordingly, we have carried out a phase I study evaluating the safety of intracerebral administration of this protein in subjects with HD, using a device formed by a semipermeable membrane encapsulating a BHK cell line engineered to synthesize CNTF. Six subjects with stage 1 or 2 HD had one capsule implanted into the right lateral ventricle; the capsule was retrieved and exchanged for a new one every 6 months, over a total period of 2 years. No sign of CNTF-induced toxicity was observed; however, depression occurred in three subjects after removal of the last capsule, which may have correlated with the lack of any future therapeutic option. All retrieved capsules were intact but contained variable numbers of surviving cells, and CNTF release was low in 13 of 24 cases. Improvements in electrophysiological results were observed, and were correlated with capsules releasing the largest amount of CNTF. This phase I study shows the safety, feasibility, and tolerability of this gene therapy procedure. Heterogeneous cell survival, however, stresses the need for improving the technique.


Assuntos
Terapia Genética/métodos , Doença de Huntington/genética , Doença de Huntington/terapia , Fármacos Neuroprotetores/farmacologia , Animais , Encéfalo/metabolismo , Linhagem Celular , Sobrevivência Celular , Fator Neurotrófico Ciliar/química , Fator Neurotrófico Ciliar/genética , Códon , Cricetinae , Eletrofisiologia , Feminino , Técnicas de Transferência de Genes , Humanos , Masculino , Neurônios/metabolismo , Polímeros/química , Retroviridae/genética , Fatores de Tempo
14.
Neurobiol Dis ; 17(2): 283-9, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15474365

RESUMO

Viral delivery of glial cell line-derived neurotrophic factor (GDNF) currently represents one of the most promising neuroprotective strategies for Parkinson's Disease (PD). However, the effect of this neurotrophic factor has never been tested in the newly available genetic models of PD based on the viral expression of mutated alpha-synuclein. In this study, we evaluated the ability of lentiviral vectors coding for GDNF (lenti-GDNF) to prevent nigral dopaminergic degeneration associated with the lentiviral mediated expression of the A30P mutant human alpha-synuclein (lenti-A30P). This virally based rat model develops a progressive and selective loss of dopamine neurons associated with the appearance of alpha-synuclein containing inclusions, thus recapitulating the major hallmarks of PD. Lenti-GDNF was injected in the substantia nigra 2 weeks before nigral administration of lenti-A30P. Although a robust expression of GDNF was observed in the whole nigrostriatal pathway due to retrograde and/or anterograde transport, lenti-GDNF did not prevent the alpha-synuclein-induced dopaminergic neurodegeneration in the lentiviral-based genetic rat model of PD. These results suggest that sustained GDNF treatment cannot modulate the cellular toxicity related to abnormal folded protein accumulation as mutated human alpha-synuclein.


Assuntos
Técnicas de Transferência de Genes , Lentivirus/genética , Degeneração Neural/etiologia , Degeneração Neural/prevenção & controle , Fatores de Crescimento Neural/farmacologia , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/complicações , Animais , Feminino , Vetores Genéticos , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Humanos , Imuno-Histoquímica , Fatores de Crescimento Neural/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Doença de Parkinson/etiologia , Doença de Parkinson/patologia , Ratos , Ratos Wistar , Sinucleínas , alfa-Sinucleína
16.
Hum Gene Ther ; 15(7): 669-80, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15242527

RESUMO

Allogeneic cells are the most attractive source for cell transplantation, as the use of xenogeneic cells is hampered by safety concerns and the use of autologous cells involves practical difficulties. The immune rejection of allogeneic cells can be overcome by physical immunoprotection provided by polymer encapsulation. To study the variability of cell and donor sources, we compared different primary human cells as candidates for gene therapy-mediated delivery of human erythropoietin (hEpo). DARC-3.1 fibroblasts, MDX-01 fibroblasts, and ARPE-19 retinal pigment epithelial cells were encapsulated into polyethersulfone hollow fibers and implanted for 1 month in nude mice as well as in immunocompetent and FK506-immunosuppressed mice to test their in vivo resistance, with the assumption that xenogeneic conditions constitute a stringent model for human application. DARC-3.1 fibroblasts showed the best survival, prompting us to evaluate cell lineages from the same donor (DARC-3.2) or another donor (DARC-4.3 and DARC-4.4). With the exception of DARC-4.3, the remaining three lineages showed comparable survival in immunocompetent C3H and DBA/2J mice. DARC-3.1 fibroblasts were retrovirally engineered with hEpo cDNA, reaching a secretion level of 170 IU of hEpo per 10(6) cells per day. Encapsulated DARC-3.1-hEpo cells led to significantly increased hematocrits in the various hosts and under various transplantation conditions. The present study shows that encapsulated primary human DARC-3.1 fibroblasts are able to survive under xenogeneic conditions and, once engineered with hEpo cDNA, to increase the hematocrit of transplanted mice.


Assuntos
Transplante de Células/métodos , Eritropoetina/genética , Fibroblastos/transplante , Animais , Sobrevivência Celular , Eritropoetina/análise , Eritropoetina/biossíntese , Fibroblastos/citologia , Fibroblastos/metabolismo , Hematócrito , Humanos , Camundongos , Camundongos Endogâmicos DBA , Polímeros/química , Sulfonas/química , Transplante Heterólogo
17.
Biomaterials ; 25(17): 3861-8, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15020162

RESUMO

The encapsulation of genetically modified cells represents a promising approach for the delivery of therapeutic proteins. The functionality of the device is dependent on the characteristics of the biomaterials, the procedures used in its confection and the adaptability of the encapsulated cells in the host. We report conditions leading to the development of calcifications on the polyvinyl alcohol (PVA) matrix introduced in hollow fiber devices for the encapsulation of primary human fibroblasts implanted in mice. The manufacturing procedures, batches of PVA matrix and cell lineages were assessed for their respective role in the development of the phenomenon. The results showed that the calcification is totally prevented by substituting phosphate-buffer saline with ultra-pure sterile water in the rinsing procedure of the matrix. Moreover, a positive correlation was found, when comparing two fibroblast cell lineages, between the level of lactate dehydrogenase (LDH) activity measured in the cells and the degree of calcium deposition. Higher LDH activity may decrease calcium depositions because it generates in the device a more acidic microenvironment inhibiting calcium precipitation. The present study defines optimized conditions for the encapsulation of primary human fibroblasts in order to avoid potentially detrimental calcifications and to allow long-term survival of encapsulated cells.


Assuntos
Materiais Biocompatíveis/química , Calcinose/patologia , Cálcio/química , Técnicas de Cultura de Células/métodos , Transplante de Células/métodos , Reação a Corpo Estranho/patologia , Álcool de Polivinil/química , Animais , Calcinose/etiologia , Linhagem Celular , Reação a Corpo Estranho/etiologia , Humanos , Masculino , Teste de Materiais , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Álcool de Polivinil/efeitos adversos
18.
Acta Neurochir Suppl ; 88: 137-41, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14531571

RESUMO

Despite aggressive surgery and post-operative radiation and chemotherapy, the prognosis is poor for glioblastoma patients. Anti-angiogenic therapy with compounds such as endostatin could delay the onset of relapse. However, the short systemic half-life of this proteins as well as the blood-brain barrier makes the use of this therapy difficult for brain cancer patients. The aim of this project is to develop and implant genetically engineered producer cells secreting endostatin that are encapsulated in calcium cross-linked alginate gel beads. Encapsulation of cells within alginate gels has a potential as a sustained release system in addition to the fact that the encapsulation technology protects the cells from rejection by the immune system. Human embryonal kidney 293 cells have been transfected with the gene for endostatin. These cells have been encapsulated in calcium cross-linked alginate gels and optimized for the secretion of endostatin. Alginate gel beads implanted into rat brain have shown only a moderate loss in cell viability but extended endostatin release for periods of up to 12 months. Visualization of the anti-angiogenic effect on C6 rat glioma growth, tumor vasculature and microhemodynamics has been demonstrated by using intravital video microscopy. The data indicates that endostatin greatly affects tumor-associated microcirculation but does not appear to affect normal microcirculation. The local delivery of endostatin seems to specifically affect tumor-associated microvessels by reduction of the vessel density, diameter and functionality. Tumor cell migration and invasion was greatly reduced in the endostatin treated animals.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Transplante de Células , Endostatinas/administração & dosagem , Marcação de Genes , Glioma/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Alginatos , Inibidores da Angiogênese/genética , Inibidores da Angiogênese/metabolismo , Animais , Neoplasias Encefálicas/irrigação sanguínea , Cápsulas , Linhagem Celular , Sistemas de Liberação de Medicamentos , Endostatinas/genética , Endostatinas/metabolismo , Glioma/irrigação sanguínea , Humanos , Microcirculação/efeitos dos fármacos , Ratos , Transfecção
19.
Gene Ther ; 10(9): 818-21, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12704422

RESUMO

For most retinal degeneration disorders, no efficient treatment exists to preserve photoreceptors (PRs) and, consequently, to maintain vision. Gene transfer appears to be a promising approach to prevent PR loss. In order to design adequate vectors to target specific retinal cell types, we have analyzed the expression pattern of three different promoters (mouse phosphoglycerate kinase 1 (PGK), elongation factor-1 (EFS), rhodopsin (Rho)) in newborn and adult DBA/2 mice retinas using self-inactivating lentiviral vectors. At 7 days after intraocular injection and in optimal conditions, cell transduction was observed up to 1.5 mm from the injection site. PGK promoter expression was predominant in the retinal pigment epithelium (RPE), especially in adult mice, whereas the EFS promoter allowed a broad expression in the retina. Finally, as expected, the Rho promoter was specifically expressed in PRs. Differences in the cell types transduced and in transduction efficiency were observed between newborn and adult injected eyes emphasizing the importance of such basic studies for further gene therapy approaches as well as for understanding the transcriptional changes during retinal maturation. Thus, for future attempts to slow or rescue retinal degeneration by lentiviral delivery, PGK and EFS are more suitable to control the expression of a supporting secreted factor, PGK being mainly expressed in RPE and EFS in different cell types throughout the entire retina, whereas Rho should allow to specifically deliver the therapeutic gene to PRs.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Lentivirus/genética , Regiões Promotoras Genéticas/genética , Degeneração Retiniana/terapia , Animais , Animais Recém-Nascidos , Expressão Gênica , Vetores Genéticos/genética , Lentivirus/fisiologia , Camundongos , Camundongos Endogâmicos DBA , Microscopia de Fluorescência , Fator 1 de Elongação de Peptídeos/genética , Fosfoglicerato Quinase/genética , Retina/metabolismo , Rodopsina/genética , Inativação de Vírus
20.
J Gene Med ; 5(3): 246-57, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12666190

RESUMO

BACKGROUND: The transplantation of encapsulated cells genetically engineered to secrete human erythropoietin (hEpo) represents an alternative to repeated injections of the recombinant hormone for the treatment of Epo-responsive anemia. In the present study, the ability of primary human foreskin fibroblasts to secrete high levels of hEpo and the importance of cis-acting elements and infection conditions on transgene expression level were assessed. METHODS: The transduction efficiency was first evaluated with beta-galactosidase (LacZ)-encoding retroviral vectors derived from the murine leukemia retrovirus (MLV) pseudotyped either with an amphotropic envelope or with the G glycoprotein of vesicular stomatitis virus (VSV-G). Human fibroblasts were then infected with an amphotropic hEpo-expressing retroviral vector, which was modified by insertion of a post-transcriptional regulatory element from the woodchuck hepatitis virus (WPRE) and a Kozak consensus sequence (KZ). Human Epo production was further optimized by increasing the multiplicity of infection and by selecting high producer cells. The survival and the transgene expression of these fibroblasts were finally evaluated in vivo. The cells were encapsulated into microporous hollow fibers and subcutaneously implanted in nude mice. RESULTS: A secretion level of approximately 5 IU hEpo/10(6) cells/day was obtained with the basal vector. A 7.5-fold increase in transgene expression was observed with the insertion of WPRE and KZ elements. Finally, according to the optimization of infection conditions, we obtained a 40-fold increase in hEpo secretion, reaching approximately 200 IU hEpo/10(6) cells/day. The in vivo experiments showed an increase in the hematocrit during the first 2 weeks and elevated levels exceeding 60% were maintained over a 6-week period. CONCLUSIONS: These results indicate that primary human fibroblasts represent a promising source for encapsulated cell therapy.


Assuntos
Eritropoetina/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Vírus da Leucemia Murina , Animais , Eritropoetina/metabolismo , Fibroblastos/metabolismo , Fibroblastos/transplante , Genes Reguladores , Genes Reporter , Humanos , Camundongos , Camundongos Nus , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...