Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Brain ; 130(Pt 3): 799-815, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17303591

ABSTRACT

Overexpression of human alpha-synuclein (alpha-syn) using recombinant adeno-associated viral (rAAV) vectors provides a novel tool to study neurodegenerative processes seen in Parkinson's disease and other synucleinopathies. We used a pseudotyped rAAV2/5 vector to express human wild-type (wt) alpha-syn, A53T mutated alpha-syn, or the green fluorescent protein (GFP) in the primate ventral midbrain. Twenty-four adult common marmosets (Callithrix jacchus) were followed with regular behavioural tests for 1 year after transduction. alpha-Syn overexpression affected motor behaviour such that all animals remained asymptomatic for at least 9 weeks, then motor bias comprising head position bias and full body rotations were seen in wt-alpha-syn expressing animals between 15 and 27 weeks; in the later phase, the animals overexpressing the A53T alpha -syn, in particular, showed a gradual worsening of motor performance, with increased motor coordination errors. Histological analysis from animals overexpressing either the wt or A53T alpha -syn showed prominent degeneration of dopaminergic fibres in the striatum. In the ventral midbrain, however, the dopaminergic neurodegeneration was more prominent in the A53T group than in the WT group suggesting differential toxicity of these two proteins in the primate brain. The surviving cell bodies and their processes in the substantia nigra were stained by antibodies to the pathological form of alpha-syn that is phosphorylated at Ser position 129. Moreover, we found, for the first time, ubiquitin containing aggregates after overexpression of alpha-syn in the primate midbrain. There was also a variable loss of oligodendroglial cells in the cerebral peduncle. These histological and behavioural data suggest that this model provides unique opportunities to study progressive neurodegeneration in the dopaminergic system and deposition of alpha-syn and ubiquitin similar to that seen in Parkinson's disease, and to test novel therapeutic targets for neuroprotective strategies.


Subject(s)
Mesencephalon/chemistry , alpha-Synuclein/analysis , Animals , Behavior, Animal/physiology , Callithrix , Dependovirus/genetics , Female , Gene Expression Regulation/genetics , Genetic Vectors , Green Fluorescent Proteins/analysis , Head/physiology , Immunohistochemistry/methods , Magnetic Resonance Imaging/methods , Male , Mesencephalon/pathology , Motor Activity/physiology , Movement/physiology , Mutation/genetics , Neuroglia/physiology , Neurons/physiology , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Recombination, Genetic , Rotation , alpha-Synuclein/genetics
2.
Neurosci Lett ; 408(2): 89-93, 2006 Nov 13.
Article in English | MEDLINE | ID: mdl-16982144

ABSTRACT

Nogo-A is a myelin-associated protein that has been shown to inhibit axonal sprouting after lesions to the CNS. Several studies have demonstrated that blocking the activity or expression of this inhibitor can induce structural and functional recovery after CNS lesions. However, there are limited and contradictory data on the expression of Nogo-A after CNS lesions. In the present study, marmoset monkeys received permanent occlusion of the middle cerebral artery (MCAo). Two, 3, or 4 months after the onset of injury brain sections were stained for Nogo-A protein. Two sham operated marmosets were included as a control. Nogo-A protein expression was quantified in white matter and grey matter in the areas adjacent to the lesion (or the equivalent areas in the intact side). At 2 months after injury, but not at 3 or 4 months, there was a significant increase in the number of oligodendrocytes that were Nogo-A immunopositive. This increase was observed in white matter structures that were adjacent to the lesion (e.g. corona radiate (CR)); but not in: white matter structures distal to the lesion (e.g. corpus callosum (CC)); cortical regions adjacent to the lesion; contralateral regions or in sham operated marmosets. These data suggest that Nogo-A levels are significantly increased within oligodendrocytes in areas adjacent to the lesion up to 2 months following cerebral ischaemia. Future studies will determine whether this offers the opportunity to promote plasticity by targeting Nogo-A weeks or months following stroke.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Growth Inhibitors/metabolism , Myelin Proteins/metabolism , Animals , Brain/cytology , Brain/pathology , Callithrix , Infarction, Middle Cerebral Artery , Nogo Proteins , Time Factors
3.
Brain Res Bull ; 68(3): 140-9, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16325013

ABSTRACT

Parkinson's disease (PD) is a debilitating neurodegenerative disease, with clinical features of tremor, muscular rigidity and akinesia, occurring as a result of midbrain dopamine loss. The search for treatments has relied heavily on animal models of the disorder. The use of monkey models of PD plays a distinct role in the development and assessment of novel treatments. The common marmoset (Callithrix jacchus) is a popular New World monkey used in the search for new treatments. These monkeys are easy to handle and survive well in captivity. This review examines the advantages of using marmoset monkeys in PD research and examines the different models available with reference to their use in pre-clinical assessment for novel therapeutic treatments. The most common models involve the administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or 6-hydroxydopamine (6-OHDA). Recently, selective cerebral transgenic over-expression of alpha-synuclein has also been attempted in marmosets as a potential model for PD. Each model has its advantages. The MPTP-based model in marmosets resembles the disease with regards to the neuroanatomy of neurotransmitter loss; the unilateral application of 6-OHDA allows for the assessment of more complex sensorimotor deficits due to the presence of an intact 'control' side; the over-expression of alpha-synuclein in the midbrain results in the slow onset of behavioural symptoms allowing for a pre-symptomatic time window. The appropriateness of each of these marmoset models for the assessment of treatments depends on several factors including the experimental aim of the study and whether emphasis is placed on the analysis of behavioural deficits.


Subject(s)
Callithrix , Disease Models, Animal , Animals , Humans , Oxidopamine/toxicity , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/physiopathology , alpha-Synuclein/metabolism
4.
J Neurosci ; 25(4): 769-77, 2005 Jan 26.
Article in English | MEDLINE | ID: mdl-15673656

ABSTRACT

The therapeutic potential of glial cell line-derived neurotrophic factor (GDNF) for Parkinson's disease is likely to depend on sustained delivery of the appropriate amount to the target areas. Recombinant adeno-associated viral vectors (rAAVs) expressing GDNF may be a suitable delivery system for this purpose. The aim of this study was to define a sustained level of GDNF that does not affect the function of the normal dopamine (DA) neurons but does provide anatomical and behavioral protection against an intrastriatal 6-hydroxydopamine (6-OHDA) lesion in the common marmoset. We found that unilateral intrastriatal injection of rAAV resulting in the expression of high levels of GDNF (14 ng/mg of tissue) in the striatum induced a substantial bilateral increase in tyrosine hydroxylase protein levels and activity as well as in DA turnover. Expression of low levels of GDNF (0.04 ng/mg of tissue), on the other hand, produced only minimal effects on DA synthesis and only on the injected side. In addition, the low level of GDNF provided approximately 85% protection of the nigral DA neurons and their projections to the striatum in the 6-OHDA-lesioned hemisphere. Furthermore, the anatomical protection was accompanied by a complete attenuation of sensorimotor neglect, head position bias, and amphetamine-induced rotation. We conclude that when delivered continuously, a low level of GDNF in the striatum (approximately threefold above baseline) is sufficient to provide optimal functional outcome.


Subject(s)
Genetic Therapy , Neostriatum/metabolism , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/genetics , Parkinson Disease/therapy , Substantia Nigra/metabolism , Animals , Appetitive Behavior/physiology , Callithrix , Choice Behavior/physiology , Dependovirus , Disease Models, Animal , Dopamine/metabolism , Female , Genetic Vectors , Glial Cell Line-Derived Neurotrophic Factor , Green Fluorescent Proteins/biosynthesis , Male , Motor Activity/physiology , Neostriatum/pathology , Nerve Growth Factors/biosynthesis , Nerve Regeneration/physiology , Oxidopamine , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Parkinson Disease, Secondary , Perceptual Disorders/physiopathology , Recombinant Fusion Proteins/biosynthesis , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
5.
Rev Neurosci ; 16(4): 303-10, 2005.
Article in English | MEDLINE | ID: mdl-16519007

ABSTRACT

Potential future treatments for Parkinson's disease (PD) include those that not only provide symptomatic relief to patients but are also neuroprotective and/or neurorestorative. Glial cell line-derived neurotrophic factor (GDNF) may be a valuable candidate in this regard. Positive results using monkeys have encouraged the use of GDNF in human trials. These trials have unfortunately shown mixed results, illustrating the influence that various parameters of administration can have on clinical outcome. The aim of this review is to compare the findings of these clinical studies with available primate data. While bolus intraventricular injections of GDNF in primates have shown some behavioural efficacy, there was no clinical benefit in the first human trial using this method, which was most likely a result of inefficient GDNF distribution in the striatal parenchyma. In primates, however, continuous (rather than bolus) delivery of GDNF into the ventricles results in significant distribution in the striatum. While chronic delivery of GDNF into the ventricles has not been assessed in humans, intraputamenal protein delivery in two Phase I trials have demonstrated that GDNF considerably reduces PD symptoms, suggesting that the putamen is the optimal location for delivery. Primate studies have shown that vector mediated delivery of GDNF may provide a suitable means for long-term intraputamenal delivery. However, the possibility of high levels of GDNF resulting in widespread distribution of GDNF to non-targeted areas is a cause of concern, and vectors with transgene regulation are needed. The development of these vectors may be the way forward for GDNF treatment.


Subject(s)
Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Parkinson Disease/drug therapy , Animals , Clinical Trials, Phase I as Topic , Genetic Therapy , Glial Cell Line-Derived Neurotrophic Factor/administration & dosage , Glial Cell Line-Derived Neurotrophic Factor/genetics , Haplorhini , Humans , Parkinson Disease/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use
6.
Exp Neurol ; 184(1): 536-48, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14637123

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) has shown potential as a treatment for Parkinson's disease. Recombinant adeno-associated viral vectors expressing the GDNF protein (rAAV-GDNF) have been used in rodent models of Parkinson's disease to promote functional regeneration after 6-OHDA lesions of the nigrostriatal system. The goal of the present study was to assess the anatomical and functional efficacy of rAAV-GDNF in the common marmoset monkey (Callithrix jacchus). rAAV-GDNF was injected into the striatum and substantia nigra 4 weeks prior to a unilateral 6-OHDA lesion of the nigrostriatal bundle. Forty percent of the dopamine cells in the lesioned substantia nigra of the rAAV-GDNF-treated monkeys survived, compared with 21% in the untreated monkeys. Fine dopaminergic fibres were observed microscopically in the injected striatum of some rAAV-GDNF-treated monkeys, suggesting that rAAV-GDNF treatment may have prevented, at least in part, the loss of dopaminergic innervation of the striatum. Protection of dopamine cells and striatal fibre innervation was associated with amelioration of the lesion-induced behavioural deficits. rAAV-GDNF-treated monkeys showed partial or complete protection not only in the amphetamine and apomorphine rotation but also in head position and the parkinsonian disability rating scale. Therefore, our study provides evidence for the behavioural and anatomical efficacy of GDNF delivered via an rAAV vector as a possible treatment for Parkinson's disease.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Membrane Transport Proteins , Nerve Growth Factors/genetics , Nerve Growth Factors/physiology , Neuropeptides , Oxidopamine/toxicity , Sympatholytics/toxicity , Animals , Body Weight/drug effects , Callithrix , Cell Count , Glial Cell Line-Derived Neurotrophic Factor , Head Movements/physiology , Locomotion/drug effects , Locomotion/physiology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Nerve Fibers/pathology , Rotation , Stereotyped Behavior/physiology , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism , Vesicular Biogenic Amine Transport Proteins
7.
Exp Neurol ; 183(2): 418-29, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14552882

ABSTRACT

Animal studies investigating the efficacy of neurotrophic factors as treatments for Parkinson's disease (PD) ideally require partial dopamine (DA) lesion models. The intrastriatal 6-hydroxydopamine (6-OHDA) lesion model may be suitable for this purpose. Although this model has been well characterized in rodents, it has not previously been used in monkeys. The goal of the present study was to characterize the behavioral effects of unilateral injections of 6-OHDA in the basal ganglia of common marmoset monkeys (Callithrix jacchus). Cell counts from tyrosine hydroxylase immunochemistry 5 months postlesion revealed DA cell loss in the substantia nigra on the lesioned side to approximately 46% of relative to the unlesioned side. 6-OHDA lesioned monkeys showed a variety of behavioral deficits. Apomorphine induced rotation and simple sensorimotor measures (head position bias and PD disability rating score) were most affected by the lesion. The largest deficits were seen at 1 or 2 weeks postsurgery but had recovered by week 10. 6-OHDA lesioned monkeys took longer to complete a more complex sensorimotor staircase task. At 3.5 months postlesion, 6-OHDA monkeys also showed deficits on an object retrieval task designed to measure sensorimotor planning and skilled hand use. alpha-Methyl-p-tyrosine, a tyrosine hydroxylase inhibitor, reinstated those deficits which had undergone recovery in the lesioned animals and also exacerbated the deficits on the staircase task. This model has potential in assessing treatments for PD aimed at curtailing disease progression such as continuous delivery of neurotrophic factors.


Subject(s)
Callithrix , Corpus Striatum/physiopathology , Disease Models, Animal , Oxidopamine , Parkinsonian Disorders/physiopathology , Animals , Behavior, Animal/drug effects , Cell Count , Corpus Striatum/drug effects , Corpus Striatum/pathology , Disability Evaluation , Disease Progression , Female , Male , Motor Activity/drug effects , Motor Skills/drug effects , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Posture , Psychomotor Performance/drug effects , Tyrosine 3-Monooxygenase/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...