Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
PLoS One ; 16(1): e0245171, 2021.
Article in English | MEDLINE | ID: mdl-33493177

ABSTRACT

Infection with Influenza A virus can lead to the development of encephalitis and subsequent neurological deficits ranging from headaches to neurodegeneration. Post-encephalitic parkinsonism has been reported in surviving patients of H1N1 infections, but not all cases of encephalitic H1N1 infection present with these neurological symptoms, suggesting that interactions with an environmental neurotoxin could promote more severe neurological damage. The heavy metal, manganese (Mn), is a potential interacting factor with H1N1 because excessive exposure early in life can induce long-lasting effects on neurological function through inflammatory activation of glial cells. In the current study, we used a two-hit model of neurotoxin-pathogen exposure to examine whether exposure to Mn during juvenile development would induce a more severe neuropathological response following infection with H1N1 in adulthood. To test this hypothesis, C57BL/6 mice were exposed to MnCl2 in drinking water (50 mg/kg/day) for 30 days from days 21-51 postnatal, then infected intranasally with H1N1 three weeks later. Analyses of dopaminergic neurons, microglia and astrocytes in basal ganglia indicated that although there was no significant loss of dopaminergic neurons within the substantia nigra pars compacta, there was more pronounced activation of microglia and astrocytes in animals sequentially exposed to Mn and H1N1, as well as altered patterns of histone acetylation. Whole transcriptome Next Generation Sequencing (RNASeq) analysis was performed on the substantia nigra and revealed unique patterns of gene expression in the dual-exposed group, including genes involved in antioxidant activation, mitophagy and neurodegeneration. Taken together, these results suggest that exposure to elevated levels of Mn during juvenile development could sensitize glial cells to more severe neuro-immune responses to influenza infection later in life through persistent epigenetic changes.


Subject(s)
Gene Expression Regulation , Influenza A Virus, H1N1 Subtype/metabolism , Manganese/pharmacology , Meningitis, Viral/metabolism , Neuroglia/metabolism , Orthomyxoviridae Infections/metabolism , Substantia Nigra/metabolism , Animals , Female , Male , Meningitis, Viral/pathology , Mice , Neuroglia/pathology , Neuroglia/virology , Orthomyxoviridae Infections/pathology , RNA-Seq , Substantia Nigra/pathology , Substantia Nigra/virology
2.
J Neurovirol ; 24(4): 398-410, 2018 08.
Article in English | MEDLINE | ID: mdl-29594983

ABSTRACT

The widespread use of antiretroviral therapy for treatment of human immunodeficiency virus (HIV) infections has dramatically improved the quality and duration of life for HIV-positive individuals. Despite this success, HIV persists for the life of an infected person in tissue reservoirs including the nervous system. Thus, whether HIV exacerbates age-related brain disorders such as Parkinson's disease (PD) is of concern. In support of this idea, HIV infection can be associated with motor and gait abnormalities that parallel late-stage manifestations of PD including dopaminergic neuronal loss. With these findings in hand, we investigated whether viral infection could affect nigrostriatal degeneration or exacerbate chemically induced nigral degeneration. We now demonstrate an additive effect of EcoHIV on dopaminergic neuronal loss and neuroinflammation induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine intoxication. HIV-1-infected humanized mice failed to recapitulate these EcoHIV results suggesting species-specific neural signaling. The results demonstrate a previously undefined EcoHIV-associated neurodegenerative response that may be used to model pathobiological aspects of PD.


Subject(s)
HIV Infections/complications , MPTP Poisoning/complications , Substantia Nigra/pathology , Substantia Nigra/virology , Animals , HIV Infections/pathology , HIV-1 , Humans , MPTP Poisoning/pathology , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced , Nerve Degeneration/virology
3.
Gene Ther ; 23(6): 520-6, 2016 06.
Article in English | MEDLINE | ID: mdl-26953486

ABSTRACT

A pilot study in nonhuman primates was conducted, in which two Rhesus macaques received bilateral parenchymal infusions of adeno-associated virus serotype 9 encoding green fluorescent protein (AAV9-GFP) into each putamen. The post-surgical in-life was restricted to 3 weeks in order to minimize immunotoxicity expected to arise from expression of GFP in antigen-presenting cells. Three main findings emerged from this work. First, the volume over which AAV9 expression was distributed (Ve) was substantially greater than the volume of distribution of MRI signal (Vd). This stands in contrast with Ve/Vd ratio of rAAV2, which is lower under similar conditions. Second, post-mortem analysis revealed expression of GFP in thalamic and cortical neurons as well as dopaminergic neurons projecting from substantia nigra pars compacta, indicating retrograde transport of AAV9. However, fibers in the substantia nigra pars reticulata, a region that receives projections from putamen, also stained for GFP, indicating anterograde transport of AAV9 as well. Finally, one hemisphere received a 10-fold lower dose of vector compared with the contralateral hemisphere (1.5 × 10(13) vg ml(-1)) and we observed a much stronger dose effect on anterograde-linked than on retrograde-linked structures. These data suggest that AAV9 can be axonally transported bi-directionally in the primate brain. This has obvious implications to the clinical developing of therapies for neurological disorders like Huntington's or Alzheimer's diseases.


Subject(s)
Axonal Transport/physiology , Brain/virology , Dependovirus/metabolism , Genetic Therapy/methods , Transduction, Genetic/methods , Animals , Antigen-Presenting Cells/metabolism , Astrocytes/metabolism , Astrocytes/virology , Axonal Transport/genetics , Brain/metabolism , Corpus Striatum/metabolism , Corpus Striatum/virology , Dependovirus/genetics , Genetic Vectors/genetics , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Macaca mulatta , Microglia/metabolism , Microglia/virology , Neurons/metabolism , Neurons/virology , Pilot Projects , Putamen/metabolism , Putamen/virology , Substantia Nigra/metabolism , Substantia Nigra/virology
4.
Neurobiol Dis ; 78: 100-14, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25818009

ABSTRACT

Phosphorylation of the α-synuclein (α-syn) protein at Ser129 [P(S129)-α-syn] was found to be the most abundant form in intracellular inclusions in brains from Parkinson's disease (PD) patients. This finding suggests that P(S129)-α-syn plays a central role in the pathogenesis of PD. However, it is at present unclear whether P(S129)-α-syn is pathogenic driving the neurodegenerative process. Rodent studies using neither the phosphomimics of human α-syn nor co-expression of human wild-type α-syn and kinases phosphorylating α-syn at Ser129 gave consistent results. One major concern in interpreting these findings is that human α-syn was expressed above physiological levels inducing neurodegeneration in rat nigral neurons. In order to exclude this confounding factor, we took a different approach and increased the phosphorylation level of endogenous α-syn. For this purpose, we took advantage of recombinant adeno-associated viral (rAAV) vectors to deliver polo-like kinase (PLK) 2 or PLK3 in the substantia nigra and investigated whether increased levels of P(S129)-α-syn compromised the function and survival of nigral dopaminergic neurons. Interestingly, we observed that hyperphosphorylated α-syn did not induce nigral dopaminergic cell death, as assessed at 1 and 4months. Furthermore, histological analysis did not show any accumulation of α-syn protein or formation of inclusions. Using in vivo microdialysis, we found that the only measurable functional alteration was the depolarisation-induced release of dopamine, while the in vivo synthesis rate of DOPA and dopamine baseline release remained unaltered. Taken together, our results suggest that phosphorylation of α-syn at Ser129 does not confer a toxic gain of function per se.


Subject(s)
Cell Cycle Proteins/metabolism , Dopaminergic Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism , Substantia Nigra/metabolism , alpha-Synuclein/metabolism , Animals , Brain/metabolism , Brain/virology , Cell Survival , Dependovirus , Dopamine/metabolism , Female , Genetic Vectors/adverse effects , Phosphorylation , Rats , Rats, Sprague-Dawley , Serine/metabolism , Substantia Nigra/virology
5.
Hum Gene Ther ; 24(6): 613-29, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23600720

ABSTRACT

Recombinant adeno-associated viral (AAV) vectors of serotypes 6, 8, and 9 were characterized as tools for gene delivery to dopaminergic neurons in the substantia nigra for future gene therapeutic applications in Parkinson's disease. While vectors of all three serotypes transduced nigral dopaminergic neurons with equal efficiency when directly injected to the substantia nigra, AAV6 was clearly superior to AAV8 and AAV9 for retrograde transduction of nigral neurons after striatal delivery. For sequential transduction of nigral dopaminergic neurons, the combination of AAV9 with AAV6 proved to be more powerful than AAV8 with AAV6 or repeated AAV6 administration. Surprisingly, single-stranded viral genomes persisted in nigral dopaminergic neurons within cell bodies and axon terminals in the striatum, and intact assembled AAV capsid was enriched in nuclei of nigral neurons, 4 weeks after virus injections to the substantia nigra. 6-Hydroxydopamine (6-OHDA)-induced degeneration of dopaminergic neurons in the substantia nigra reduced the number of viral genomes in the striatum, in line with viral genome persistence in axon terminals. However, 6-OHDA-induced axonal degeneration did not induce any transsynaptic spread of AAV infection in the striatum. Therefore, the potential presence of viral particles in axons may not represent an important safety issue for AAV gene therapy applications in neurodegenerative diseases.


Subject(s)
Dependovirus/metabolism , Neurons/metabolism , Neurons/virology , Substantia Nigra/metabolism , Substantia Nigra/virology , Transduction, Genetic , Virion/metabolism , Animals , Axons/metabolism , Biological Transport , Capsid/metabolism , Cell Nucleus/metabolism , Cell Nucleus/virology , Deoxyribonuclease I/metabolism , Dependovirus/genetics , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/virology , Female , Genetic Vectors , Genome, Viral/genetics , HEK293 Cells , Humans , Injections , Neostriatum/pathology , Neostriatum/virology , Nerve Degeneration/pathology , Neurons/pathology , Oxidopamine , Rats , Rats, Sprague-Dawley , Substantia Nigra/pathology
6.
PLoS One ; 6(5): e20495, 2011.
Article in English | MEDLINE | ID: mdl-21655265

ABSTRACT

Although much is known regarding the molecular mechanisms leading to neuronal cell loss in Parkinson's disease (PD), the initiating event has not been identified. Prevailing theories including a chemical insult or infectious agent have been postulated as possible triggers, leading to neuroinflammation. We present immunohistochemical data indicating the presence of influenza A virus within the substantia nigra pars compacta (SNpc) from postmortem PD brain sections. Influenza A virus labeling was identified within neuromelanin granules as well as on tissue macrophages in the SNpc. Further supporting a role for neuroinflammation in PD was the identification of T-lymphocytes that colocalized with an antibody to caspase-cleaved Beclin-1 within the SNpc. The presence of influenza A virus together with macrophages and T-lymphocytes may contribute to the neuroinflammation associated with this disease.


Subject(s)
Brain/metabolism , Brain/virology , Inflammation/metabolism , Influenza A virus/isolation & purification , Parkinson Disease/metabolism , Parkinson Disease/virology , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Humans , Immunohistochemistry , In Situ Nick-End Labeling , In Vitro Techniques , Melanins/metabolism , Membrane Proteins/metabolism , Microscopy, Fluorescence , Substantia Nigra/metabolism , Substantia Nigra/virology , T-Lymphocytes/metabolism
8.
Ann Neurol ; 70(1): 110-20, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21437936

ABSTRACT

OBJECTIVE: A prevailing concept in neuroscience has been that the adult mammalian central nervous system is incapable of restorative axon regeneration. Recent evidence, however, has suggested that reactivation of intrinsic cellular programs regulated by protein kinase B (Akt)/mammalian target of rapamycin (mTor) signaling may restore this ability. METHODS: To assess this possibility in the brain, we have examined the ability of adenoassociated virus (AAV)-mediated transduction of dopaminergic neurons of the substantia nigra (SN) with constitutively active forms of the kinase Akt and the GTPase Ras homolog enriched in brain (Rheb) to induce regrowth of axons after they have been destroyed by neurotoxin lesion. RESULTS: Both constitutively active myristoylated Akt and hRheb(S16H) induce regrowth of axons from dopaminergic neurons to their target, the striatum. Histological analysis demonstrates that these new axons achieve morphologically accurate reinnervation. In addition, functional reintegration into target circuitry is achieved, as indicated by partial behavioral recovery. INTERPRETATION: We conclude that regrowth of axons within the adult nigrostriatal projection, a system that is prominently affected in Parkinson's disease, can be achieved by activation of Akt/mTor signaling in surviving endogenous mesencephalic dopaminergic neurons by viral vector transduction.


Subject(s)
Axons/physiology , Dopamine/physiology , Monomeric GTP-Binding Proteins/administration & dosage , Nerve Regeneration/physiology , Neuropeptides/administration & dosage , Proto-Oncogene Proteins c-akt/administration & dosage , Signal Transduction , Animals , Axons/metabolism , Dependovirus/genetics , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monomeric GTP-Binding Proteins/genetics , Nerve Regeneration/genetics , Neurons/cytology , Neurons/pathology , Neurons/virology , Neuropeptides/genetics , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Proto-Oncogene Proteins c-akt/genetics , Ras Homolog Enriched in Brain Protein , Signal Transduction/genetics , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/virology
9.
J Child Neurol ; 25(4): 497-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20139405

ABSTRACT

Poliomyelitis, though eradicated from most parts of the world, continues to occur in India. There is paucity of data on the magnetic resonance imaging (MRI) changes in poliomyelitis. We report a 3(1/2)-year-old boy who presented with subacute onset flaccid paralysis and altered sensorium. Stool culture was positive for wild polio virus type 3. Magnetic resonance imaging revealed signal changes in bilateral substantia nigra and anterior horns of the spinal cord. These MRI changes may be of potential diagnostic significance in a child with poliomyelitis.


Subject(s)
Magnetic Resonance Imaging/methods , Mesencephalon/pathology , Poliomyelitis/pathology , Spinal Cord/pathology , Anterior Horn Cells/pathology , Anterior Horn Cells/virology , Child, Preschool , Comorbidity , Consciousness Disorders/pathology , Consciousness Disorders/physiopathology , Consciousness Disorders/virology , Fever/virology , Humans , India , Male , Mesencephalon/physiopathology , Mesencephalon/virology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Muscular Atrophy/virology , Paralysis/physiopathology , Paralysis/virology , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Parkinson Disease/virology , Poliomyelitis/physiopathology , Poliovirus Vaccine, Inactivated , Spinal Cord/physiopathology , Spinal Cord/virology , Substantia Nigra/pathology , Substantia Nigra/virology , Time
11.
J Neurosci ; 29(11): 3365-73, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19295143

ABSTRACT

Little is known about key pathological events preceding overt neuronal degeneration in Parkinson's disease (PD) and alpha-synucleinopathy. Recombinant adeno-associated virus 2-mediated delivery of mutant (A53T) human alpha-synuclein into the substantia nigra (SN) under a neuron-specific synapsin promoter resulted in protracted neurodegeneration with significant dopaminergic (DA) neuron loss by 17 weeks. As early as 4 weeks, there was an increase in a dopamine metabolite, DOPAC and histologically, DA axons in the striatum were dystrophic with degenerative bulbs. Before neuronal loss, significant changes were identified in levels of proteins relevant to synaptic transmission and axonal transport in the striatum and the SN. For example, striatal levels of rabphilin 3A and syntaxin were reduced. Levels of anterograde transport motor proteins (KIF1A, KIF1B, KIF2A, and KIF3A) were decreased in the striatum, whereas retrograde motor proteins (dynein, dynamitin, and dynactin1) were increased. In contrast to reduced levels in the striatum, KIF1A and KIF2A levels were elevated in the SN. There were dramatic changes in cytoskeletal protein levels, with actin levels increased and alpha-/gamma-tubulin levels reduced. In addition to these alterations, a neuroinflammatory response was observed at 8 weeks in the striatum, but not in the SN, demonstrated by increased levels of Iba-1, activated microglia and increased levels of proinflammatory cytokines, including IL-1beta, IFN-gamma and TNF-alpha. These results demonstrate that changes in proteins relevant to synaptic transmission and axonal transport coupled with neuroinflammation, precede alpha-synuclein-mediated neuronal death. These findings can provide ideas for antecedent biomarkers and presymptomatic interventions in PD.


Subject(s)
Carrier Proteins/metabolism , Dependovirus , Disease Models, Animal , Dopamine/physiology , Nervous System Diseases/metabolism , Presynaptic Terminals/metabolism , alpha-Synuclein/metabolism , Animals , Axonal Transport/genetics , Carrier Proteins/genetics , Cell Death/genetics , Dependovirus/genetics , Dopamine/genetics , Female , Genetic Vectors/genetics , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/virology , Nervous System Diseases/pathology , Nervous System Diseases/virology , Neurons/metabolism , Neurons/pathology , Neurons/virology , Presynaptic Terminals/pathology , Presynaptic Terminals/virology , Rats , Rats, Sprague-Dawley , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/virology , alpha-Synuclein/genetics
12.
J Neurochem ; 109(3): 838-45, 2009 May.
Article in English | MEDLINE | ID: mdl-19250335

ABSTRACT

Enhanced delivery and expression of genes in specific neuronal systems is critical for the development of genetic models of neurodegenerative disease and potential gene therapy. Recent discovery of new recombinant adeno-associated viral (rAAV) capsid serotypes has resulted in improved transduction efficiency, but expression levels, spread of transgene, and potential toxicity can differ depending on brain region and among species. We compared the transduction efficiency of titer-matched rAAV 2/1, 2/5, and 2/8 to the commonly used rAAV2/2 in the rat nigrostriatal system via expression of the reporter transgene, enhanced green fluorescent protein. Newer rAAV serotypes 2/1, 2/5, and 2/8 demonstrated marked increase in transduction and spread of enhanced green fluorescent protein expression in dopaminergic nigrostriatal neurons and projections to the striatum and globus pallidus compared to rAAV2/2 at 2 weeks post-injection. The number of nigral cells transduced was greatest for rAAV2/1, but for serotypes 2/5 and 2/8 was still two- to threefold higher than that for 2/2. Enhanced transduction did not cause an increase in glial cell response or toxicity. New rAAV serotypes thus promise improved gene delivery to nigrostriatal system with the potential for better models and therapeutics for Parkinson disease and other neurodegenerative disorders.


Subject(s)
Adenoviridae/classification , Adenoviridae/genetics , Genetic Vectors , Recombination, Genetic , Substantia Nigra/metabolism , Transduction, Genetic/methods , Animals , Gene Expression/physiology , Genetic Vectors/classification , Genetic Vectors/genetics , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Inflammation/virology , Rats , Rats, Sprague-Dawley , Serotyping , Substantia Nigra/virology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
13.
J Neurovirol ; 13(3): 210-24, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17613711

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) enters the central nervous system shortly after the infection and becomes localized in different regions of the brain, leading to various neurological abnormalities including motor disorders and neurocognitive deficits. Although HIV-1-associated functional abnormalities of the central nervous system (CNS) can be evaluated during life by using various test batteries, HIV-1 virus concentration in different brain regions can be measured only after death. The tissues obtained at autopsy provide a valuable source for determining the role of various factors, including that of HIV-1 viral load in the CNS, that may contribute to the regional CNS neuropathogenesis. For this study, we obtained from the National Institutes of Health-sponsored National NeuroAIDS Tissue Consortium (NNTC) the tissues from different brain regions collected at autopsy of HIV-1-positive (N = 38) and HIV-negative (N = 11) individuals, with postmortem intervals of 2 to 29 h, and measured HIV-1 RNA concentration in the frontal cortex, frontal cortex area 4, frontal cortex area 6, basal ganglia, caudate nucleus, putamen, globus pallidus, substantia nigra, and cerebrospinal fluid. Because HIV-1+ individuals were infected with the virus for up to 21 years and the majority of them had used highly active antiretroviral therapy (HAART), we used highly sensitive real-time reverse transcriptase-polymerase chain reaction (RT-PCR) assay in order to detect a wide dynamic range of HIV-1 RNA with lower detection limit of a single copy. The primers and probes were from the long terminal repeat (LTR) region of HIV genome for achieving higher specificity and sensitivity of detection and amplification. Our results demonstrate a wide variation in the concentration of HIV-1 RNA in different brain regions (5.51 and 8,144,073; log(10) 0.74 and 6.91 copies/g tissue), and despite the high specificity and sensitivity of this method, viral RNA was not detected in 50% of all the samples, and in 30% to 64% of samples of each region of HIV-1+ individuals. However, the highest concentration of viral RNA was found in the caudate nucleus and the lowest concentration in the frontal cortex and cerebrospinal fluid. The viral RNA was undetectable in all samples of HIV-negative individuals.


Subject(s)
AIDS Dementia Complex/virology , Brain/virology , HIV-1/genetics , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Adult , Aged , Caudate Nucleus/virology , Female , Frontal Lobe/virology , Globus Pallidus/virology , Humans , Male , Middle Aged , Putamen/virology , RNA, Viral/cerebrospinal fluid , Sensitivity and Specificity , Substantia Nigra/virology , Viral Load
14.
Mov Disord ; 22(8): 1124-32, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17443702

ABSTRACT

Neurturin (NTN) is a potent survival factor for midbrain dopaminergic neurons. CERE-120, an adeno-associated virus type 2 (AAV2) vector encoding human NTN (AAV2-NTN), is currently being developed as a potential therapy for Parkinson's disease. This study examined the bioactivity and safety/tolerability of AAV2-NTN in the aged monkey model of nigrostriatal dopamine insufficiency. Aged rhesus monkeys received unilateral injections of AAV2-NTN into the caudate and putamen, with each animal therefore serving as its own control. Robust expression of NTN within the nigrostriatal system was observed 8 months postadministration. (18)F-fluorodopa imaging using positron emission tomography revealed statistically significant increases in (18)F-fluorodopa uptake in the injected striatum compared with the uninjected side at 4 and 8 months. In addition, at 8 months postadministration, a significant enhancement in tyrosine hydroxylase immunoreactive fibers and an increase in the number of tyrosine hydroxylase immunoreactive cells was observed in the AAV2-NTN injected striatum compared with the uninjected side. Robust activation of phosphorylated extracellular signal-regulated kinase immunoreactivity in the substantia nigra was also observed. Histopathological analyses revealed no adverse effects of AAV2-NTN in the brain. Collectively, these results are consistent with the neurotrophic effects of NTN on the dopaminergic nigrostriatal system and extend the growing body of evidence supporting the concept that AAV2-NTN may have therapeutic benefit for Parkinson's disease.


Subject(s)
Aging/physiology , Corpus Striatum , Dependovirus/metabolism , Disease Models, Animal , Dopamine/metabolism , Neurturin/therapeutic use , Parkinson Disease , Substantia Nigra , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Corpus Striatum/virology , Female , Fluorodeoxyglucose F18/pharmacokinetics , Macaca mulatta , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/virology , Positron-Emission Tomography , Radiopharmaceuticals/pharmacokinetics , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/virology
15.
Exp Neurol ; 204(2): 791-801, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17320866

ABSTRACT

Affymetrix GeneChip technology and quantitative real-time PCR (Q-PCR) were used to examine changes in gene expression in the adult murine substantia nigra pars compacta (SNc) following lentiviral glial cell line-derived neurotrophic factor (GDNF) delivery in adult striatum. We identified several genes that were upregulated after GDNF treatment. Among these, the gene encoding the transmembrane protein Delta-like 1 homologue (Dlk1) was upregulated with a greater than 4-fold increase in mRNA encoding this protein. Immunohistochemistry with a Dlk1-specific antibody confirmed the observed upregulation with increased positive staining of cell bodies in the SNc and fibers in the striatum. Analysis of the developmental regulation of Dlk1 in the murine ventral midbrain showed that the upregulation of Dlk1 mRNA correlated with the generation of tyrosine hydroxylase (TH)-positive neurons. Furthermore, Dlk1 expression was analyzed in MesC2.10 cells, which are derived from embryonic human mesencephalon and capable of undergoing differentiation into dopaminergic neurons. We detected upregulation of Dlk1 mRNA and protein under conditions where MesC2.10 cells differentiate into a dopaminergic phenotype (41.7+/-7.1% Dlk1+ cells). In contrast, control cultures subjected to default differentiation into non-dopaminergic neurons only expressed very few (3.7+/-1.3%) Dlk1-immunopositive cells. The expression of Dlk1 in MesC2.10 cells was specifically upregulated by the addition of GDNF. Thus, our data suggest that Dlk1 expression precedes the appearance of TH in mesencephalic cells and that levels of Dlk1 are regulated by GDNF.


Subject(s)
Cell Differentiation/physiology , Dopamine/metabolism , Gene Expression Regulation/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Substantia Nigra/metabolism , Animals , Cell Differentiation/drug effects , Cell Line, Transformed , Enzyme-Linked Immunosorbent Assay/methods , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Humans , Indoles , Lentivirus/physiology , Mice , Oligonucleotide Array Sequence Analysis/methods , Substantia Nigra/virology
16.
J Neurobiol ; 66(12): 1311-21, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16967504

ABSTRACT

Uninfected neurons of the substantia nigra (SN) degenerate in human immunodeficiency virus (HIV)-positive patients through an unknown etiology. The HIV envelope glycoprotein 120 (gp120) causes apoptotic neuronal cell death in the rodent striatum, but its primary neurotoxic mechanism is still under investigation. Previous studies have shown that gp120 causes neurotoxicity in the rat striatum by reducing brain-derived neurotrophic factor (BDNF). Because glial cell line-derived neurotrophic factor (GDNF) and BDNF are neurotrophic factors crucial for the survival of dopaminergic neurons of the SN, we investigated whether gp120 reduces GDNF and BDNF levels concomitantly to induce apoptosis. Rats received a microinjection of gp120 or vehicle into the striatum and were sacrificed at various time intervals. GDNF but not BDNF immunoreactivity was decreased in the SN by 4 days in gp120-treated rats. In these animals, a significant increase in the number of caspase-3- positive neurons, both tyrosine hydroxylase (TH)-positive and -negative, was observed. Analysis of TH immunoreactivity revealed fewer TH-positive neurons and fibers in a medial and lateral portion of cell group A9 of the SN, an area that projects to the striatum, suggesting that gp120 induces retrograde degeneration of nigrostriatal neurons. We propose that dysfunction of the nigrostriatal dopaminergic system associated with HIV may be caused by a reduction of neurotrophic factor expression by gp120.


Subject(s)
Apoptosis/physiology , Brain-Derived Neurotrophic Factor/metabolism , HIV Envelope Protein gp120/metabolism , Nerve Degeneration/physiopathology , Substantia Nigra/physiopathology , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/physiopathology , Animals , Axonal Transport/physiology , Basal Ganglia Diseases/metabolism , Basal Ganglia Diseases/physiopathology , Basal Ganglia Diseases/virology , Caspase 3/metabolism , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Corpus Striatum/virology , Disease Models, Animal , Dopamine/metabolism , Down-Regulation/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , HIV Envelope Protein gp120/toxicity , HIV-1 , Male , Nerve Degeneration/metabolism , Nerve Degeneration/virology , Rats , Rats, Sprague-Dawley , Substantia Nigra/metabolism , Substantia Nigra/virology , Tyrosine 3-Monooxygenase/metabolism
17.
Brain Res ; 1095(1): 178-89, 2006 Jun 20.
Article in English | MEDLINE | ID: mdl-16729982

ABSTRACT

To produce an animal model of a dopa-responsive motor disorder with depletion of dopamine (DA) release in the striatum by dysfunction of the transmitter release machinery of the nigrostriatal DA system, we performed an intra-nigral injection of an HVJ-liposome gene transfer vector containing antisense oligodeoxynucleotides (ODNs) against synaptotagmin I (SytI), a key regulator of Ca(2+)-dependent exocytosis and endocytosis in adult rats. A unilateral intra-nigral injection of HVJ-liposome vectors containing antisense ODNs against SytI (syt-AS) caused a moderate disruption of methamphetamine-induced release of DA in the treated side of the striatum, while the syt-AS treatment did not affect physiological release of DA in the treated striatum. A bilateral intra-nigral injection of HVJ-liposome vectors containing syt-AS induced an impairment of the striatal DA-mediated acquisition of skilled behavior in a rotarod task without any deficits in general motor functions, such as spontaneous locomotor activity, motor adjusting steps, equilibrium function, or muscle strength. These findings suggest that an intra-nigral treatment with HVJ-liposome vectors containing syt-AS may cause a long-lasting nigral knockdown of SytI which, in turn, leads to a moderate dysfunction of the DA release machinery in the terminals of the nigrostriatal DA system and a subsequent mild depletion of DA release in the striatum.


Subject(s)
Corpus Striatum/metabolism , Gene Transfer Techniques , Motor Activity/physiology , Motor Skills Disorders/physiopathology , Oligonucleotides, Antisense/administration & dosage , Substantia Nigra/drug effects , Analysis of Variance , Animals , Behavior, Animal , Corpus Striatum/virology , Disease Models, Animal , Dopamine/metabolism , Immunohistochemistry/methods , Male , Methamphetamine/toxicity , Microdialysis/methods , Motor Activity/drug effects , Motor Skills/drug effects , Motor Skills/physiology , Motor Skills Disorders/etiology , Motor Skills Disorders/metabolism , Oligonucleotides, Antisense/genetics , Oxidopamine/toxicity , Rats , Rats, Wistar , Reaction Time/drug effects , Reaction Time/physiology , Rotarod Performance Test/methods , Substantia Nigra/injuries , Substantia Nigra/virology , Synaptotagmin I/deficiency
18.
Mol Ther ; 13(3): 631-5, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16423561

ABSTRACT

The Semliki Forest virus (SFV) 1 vector system is highly efficient at gene transduction in a broad range of host cells, including neurons. To determine the potential of SFV1-based vectors to mediate gene expression in substantia nigra neurons, we inoculated d1EGFP-expressing SFV virus-like particles stereotaxically into the mouse brain. This system selectively and extensively mediated gene expression in dopaminergic neurons of the substantia nigra. Continual reporter gene expression was evident in neuronal cell bodies for up to 3 weeks postinoculation and d1EGFP-positive neuronal processes were apparent for 12 weeks. There was no evidence of an apoptotic response to infection, but with time cell degeneration and an axonopathy, indicative of neuronal loss, were increasingly apparent. This system has potential for experimental studies requiring efficient transient gene transduction of mouse CNS neurons. The current SFV1 vector system is, however, limited in its potential for CNS gene therapy by neurotoxicity.


Subject(s)
Genetic Therapy/adverse effects , Genetic Vectors/toxicity , Neurons/virology , Semliki forest virus/genetics , Animals , Genetic Vectors/administration & dosage , Inflammation/virology , Mice , Neurons/pathology , Semliki forest virus/pathogenicity , Substantia Nigra/pathology , Substantia Nigra/virology
19.
Folia Morphol (Warsz) ; 64(3): 130-44, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16228947

ABSTRACT

CNS gene transfer could provide new approaches to the modelling of neurodegenerative diseases and devising potential therapies. One such disorder is Parkinson's disease (PD), in which dysfunction of several different metabolic processes has been implicated. Here we review the literature on gene transfer systems based on herpes simplex virus type 1 (HSV-1) and non-viral polyethyleneimine (PEI) and calcium phosphate nanoparticle methods. We also assess the usefulness of various CNS gene delivery methods and present some of our own data to exemplify such usefulness. Our data result from vectors stereotaxically introduced to the substantia nigra (SN) of adult rats and evaluated 1 week and/or 1 month post injection using histochemical methods to assess recombinant ss-galactosidase enzyme activity. Gene transfer using PEI or calcium phosphate-mediated transfections was observed for both methods and PEI was comparable to that of HSV-1 amplicon. Our data show that the amplicon delivery was markedly increased when packaged with a helper virus and was similar to the expression profile achieved with a full-size replication-defective HSV-1 recombinant (8117/43). We also examine whether PEI or HSV-1 amplicon-mediated gene transfer could facilitate assessment of the biological effects induced by a dominant negative FGF receptor-1 mutant to model the reduced FGF signalling thought to occur in Parkinson's disease.


Subject(s)
Calcium Phosphates/chemistry , Genetic Vectors , Herpesvirus 1, Human/genetics , Parkinson Disease/etiology , Polyethyleneimine/chemistry , Receptor Protein-Tyrosine Kinases , Receptors, Fibroblast Growth Factor , Substantia Nigra/metabolism , Animals , Animals, Newborn , Brain/metabolism , Brain/pathology , Brain/virology , Cells, Cultured , Cricetinae , Mice , NIH 3T3 Cells , Nanotechnology , Rats , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/chemistry , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Substantia Nigra/virology , Transduction, Genetic/methods
20.
Brain Res Mol Brain Res ; 139(2): 361-6, 2005 Oct 03.
Article in English | MEDLINE | ID: mdl-16039006

ABSTRACT

The effects of HSV-1 amplicon and polyethyleneimine (PEI)-mediated transfection of dominant negative FGF receptor-1 mutant FGFR1(TK-) into the rat brain substantia nigra (SN) were examined in vivo to model the reduced FGF signaling documented to occur in Parkinson's disease. The number of SN neurons that expressed tyrosine hydroxylase (TH) was significantly reduced following HSV-1 FGFR1(TK-) intranigral delivery and similar changes were observed after PEI-mediated FGFR1(TK-) transfections. Further, we also observed a significantly lower striatal dopamine content following the PEI transfection of FGFR1(TK-). Thus, we conclude that reduced FGF signaling in the SN of Parkinsonian patients could play a role in the impaired dopaminergic transmission associated with the degenerative disease.


Subject(s)
Dopamine/metabolism , Gene Expression Regulation/physiology , Neurons/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Substantia Nigra/cytology , Tyrosine 3-Monooxygenase/metabolism , Animals , Gene Expression Regulation/drug effects , Herpesvirus 1, Human/physiology , Male , Microinjections/methods , Mutagenesis/physiology , Neurons/virology , Polyethyleneimine/pharmacology , Protein-Tyrosine Kinases/deficiency , Rats , Rats, Inbred F344 , Receptor, Fibroblast Growth Factor, Type 1/genetics , Substantia Nigra/metabolism , Substantia Nigra/virology , Time Factors , Transfection/methods , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...