Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(26): e2118755119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35749364

ABSTRACT

Retromer is a heteropentameric complex that plays a specialized role in endosomal protein sorting and trafficking. Here, we report a reduction in the retromer proteins-vacuolar protein sorting 35 (VPS35), VPS26A, and VPS29-in patients with amyotrophic lateral sclerosis (ALS) and in the ALS model provided by transgenic (Tg) mice expressing the mutant superoxide dismutase-1 G93A. These changes are accompanied by a reduction of levels of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunit GluA1, a proxy of retromer function, in spinal cords from Tg SOD1G93A mice. Correction of the retromer deficit by a viral vector expressing VPS35 exacerbates the paralytic phenotype in Tg SOD1G93A mice. Conversely, lowering Vps35 levels in Tg SOD1G93A mice ameliorates the disease phenotype. In light of these findings, we propose that mild alterations in retromer inversely modulate neurodegeneration propensity in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Vesicular Transport Proteins , Amyotrophic Lateral Sclerosis/metabolism , Animals , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Spinal Cord/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
2.
Sci Rep ; 9(1): 19402, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31852909

ABSTRACT

Focused ultrasound-enhanced intranasal (IN + FUS) delivery is a noninvasive approach that utilizes the olfactory pathway to administer pharmacological agents directly to the brain, allowing for a more homogenous distribution in targeted locations compared to IN delivery alone. However, whether such a strategy has therapeutic values, especially in neurodegenerative disorders such as Parkinson's disease (PD), remains to be established. Herein, we evaluated whether the expression of tyrosine hydroxylase (TH), the rate limiting enzyme in dopamine catalysis, could be enhanced by IN + FUS delivery of brain-derived neurotrophic factor (BDNF) in a toxin-based PD mouse model. Mice were put on the subacute dosing regimen of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), producing bilateral degeneration of the nigrostriatal pathway consistent with early-stage PD. MPTP mice then received BDNF intranasally followed by multiple unilateral FUS-induced blood-brain barrier (BBB) openings in the left basal ganglia for three consecutive weeks. Subsequently, mice were survived for two months and were evaluated morphologically and behaviorally to determine the integrity of their nigrostriatal dopaminergic pathways. Mice receiving IN + FUS had significantly increased TH immunoreactivity in the treated hemisphere compared to the untreated hemisphere while mice receiving only FUS-induced BBB opening or no treatment at all did not show any differences. Additionally, behavioral changes were only observed in the IN + FUS treated mice, indicating improved motor control function in the treated hemisphere. These findings demonstrate the robustness of the method and potential of IN + FUS for the delivery of bioactive factors for treatment of neurodegenerative disorder.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Dopamine/genetics , Parkinson Disease, Secondary/therapy , Parkinson Disease/therapy , Tyrosine 3-Monooxygenase/genetics , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Administration, Intranasal , Animals , Basal Ganglia/drug effects , Basal Ganglia/pathology , Basal Ganglia/radiation effects , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/radiation effects , Brain/drug effects , Brain/pathology , Brain/radiation effects , Corpus Striatum/diagnostic imaging , Corpus Striatum/drug effects , Corpus Striatum/radiation effects , Disease Models, Animal , Dopamine/biosynthesis , Humans , Mice , Neuroprotective Agents/pharmacology , Parkinson Disease/genetics , Parkinson Disease/pathology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/pathology , Substantia Nigra/diagnostic imaging , Substantia Nigra/drug effects , Substantia Nigra/radiation effects , Ultrasonic Waves
3.
J Control Release ; 303: 289-301, 2019 06 10.
Article in English | MEDLINE | ID: mdl-30953664

ABSTRACT

The blood-brain barrier (BBB) prevents most drugs from gaining access to the brain parenchyma, which is a recognized impediment to the treatment of neurodegenerative disorders like Parkinson's disease (PD). Focused ultrasound (FUS), in conjunction with systemically administered microbubbles, opens the BBB locally, reversibly and non-invasively. Herein, we show that neither FUS applied over both the striatum and the ventral midbrain, without neurotrophic factors, nor intravenous administration of neurotrophic factors (either through protein or gene delivery) without FUS, ameliorates the damage to the nigrostriatal dopaminergic pathway in the sub-acute MPTP mouse model of early-stage PD. Conversely, the combination of FUS and intravenous neurotrophic (protein or gene) delivery attenuates the damage to the nigrostriatal dopaminergic pathway, by allowing the entry of these agents into the brain parenchyma. Our findings provide evidence that the application of FUS at the early stages of PD facilitates critical neurotrophic delivery that can curb the rapid progression of neurodegeneration while improving the neuronal function, seemingly opening new therapeutic avenues for the early treatment of diseases of the central nervous system.


Subject(s)
Genetic Therapy , Parkinsonian Disorders/therapy , Ultrasonic Therapy , Animals , Brain/metabolism , Genetic Vectors , Glial Cell Line-Derived Neurotrophic Factor/genetics , Male , Mice, Inbred C57BL , Microbubbles , Neurturin/administration & dosage , Recombinant Proteins/administration & dosage
4.
Front Neuroanat ; 9: 91, 2015.
Article in English | MEDLINE | ID: mdl-26217195

ABSTRACT

Parkinson disease (PD) is a chronic, progressive neurological disease that is associated with a loss of dopaminergic neurons in the substantia nigra pars compacta of the brain. The molecular mechanisms underlying the loss of these neurons still remain elusive. Oxidative stress is thought to play an important role in dopaminergic neurotoxicity. Complex I deficiencies of the respiratory chain account for the majority of unfavorable neuronal degeneration in PD. Environmental factors, such as neurotoxins, pesticides, insecticides, dopamine (DA) itself, and genetic mutations in PD-associated proteins contribute to mitochondrial dysfunction which precedes reactive oxygen species formation. In this mini review, we give an update of the classical pathways involving these mechanisms of neurodegeneration, the biochemical and molecular events that mediate or regulate DA neuronal vulnerability, and the role of PD-related gene products in modulating cellular responses to oxidative stress in the course of the neurodegenerative process.

5.
Nat Neurosci ; 18(9): 1325-33, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26214373

ABSTRACT

For degenerative disorders of the CNS, the main obstacle to therapeutic advancement has been the challenge of identifying the key molecular mechanisms underlying neuronal loss. We developed a combinatorial approach including translational profiling and brain regulatory network analysis to search for key determinants of neuronal survival or death. Following the generation of transgenic mice for cell type-specific profiling of midbrain dopaminergic neurons, we established and compared translatome libraries reflecting the molecular signature of these cells at baseline or under degenerative stress. Analysis of these libraries by interrogating a context-specific brain regulatory network led to the identification of a repertoire of intrinsic upstream regulators that drive the dopaminergic stress response. The altered activity of these regulators was not associated with changes in their expression levels. This strategy can be generalized for the identification of molecular determinants involved in the degeneration of other classes of neurons.


Subject(s)
Dopaminergic Neurons/metabolism , Mesencephalon/metabolism , Nerve Net/metabolism , Neurodegenerative Diseases/metabolism , Protein Biosynthesis/physiology , Substantia Nigra/metabolism , Animals , Dopaminergic Neurons/pathology , Male , Mesencephalon/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Substantia Nigra/pathology
6.
J Neurosci ; 35(14): 5724-42, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25855184

ABSTRACT

Accumulating evidence from genetic and biochemical studies implicates dysfunction of the autophagic-lysosomal pathway as a key feature in the pathogenesis of Parkinson's disease (PD). Most studies have focused on accumulation of neurotoxic α-synuclein secondary to defects in autophagy as the cause of neurodegeneration, but abnormalities of the autophagic-lysosomal system likely mediate toxicity through multiple mechanisms. To further explore how endolysosomal dysfunction causes PD-related neurodegeneration, we generated a murine model of Kufor-Rakeb syndrome (KRS), characterized by early-onset Parkinsonism with additional neurological features. KRS is caused by recessive loss-of-function mutations in the ATP13A2 gene encoding the endolysosomal ATPase ATP13A2. We show that loss of ATP13A2 causes a specific protein trafficking defect, and that Atp13a2 null mice develop age-related motor dysfunction that is preceded by neuropathological changes, including gliosis, accumulation of ubiquitinated protein aggregates, lipofuscinosis, and endolysosomal abnormalities. Contrary to predictions from in vitro data, in vivo mouse genetic studies demonstrate that these phenotypes are α-synuclein independent. Our findings indicate that endolysosomal dysfunction and abnormalities of α-synuclein homeostasis are not synonymous, even in the context of an endolysosomal genetic defect linked to Parkinsonism, and highlight the presence of α-synuclein-independent neurotoxicity consequent to endolysosomal dysfunction.


Subject(s)
Adenosine Triphosphatases/deficiency , Brain/metabolism , Lysosomes/metabolism , Membrane Proteins/deficiency , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , alpha-Synuclein/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/ultrastructure , Animals , Brain/pathology , Brain/ultrastructure , Cytosol/metabolism , Cytosol/ultrastructure , Disease Models, Animal , Dopaminergic Neurons/pathology , Endosomes/metabolism , Endosomes/ultrastructure , Exploratory Behavior/physiology , Hindlimb Suspension/psychology , Hydrogen-Ion Concentration , Lipids/analysis , Lysosomes/ultrastructure , Male , Membrane Proteins/genetics , Membrane Proteins/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Nerve Tissue Proteins/metabolism , Parkinsonian Disorders/physiopathology , Postural Balance/genetics , Proton-Translocating ATPases
7.
Neuron ; 75(2): 306-19, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22841315

ABSTRACT

Non cell-autonomous processes are thought to play critical roles in the cellular maintenance of the healthy and diseased brain but mechanistic details remain unclear. We report that the interruption of a non cell-autonomous mode of sonic hedgehog (Shh) signaling originating from dopaminergic neurons causes progressive, adult-onset degeneration of dopaminergic, cholinergic, and fast spiking GABAergic neurons of the mesostriatal circuit, imbalance of cholinergic and dopaminergic neurotransmission, and motor deficits reminiscent of Parkinson's disease. Variable Shh signaling results in graded inhibition of muscarinic autoreceptor- and glial cell line-derived neurotrophic factor (GDNF)-expression in the striatum. Reciprocally, graded signals that emanate from striatal cholinergic neurons and engage the canonical GDNF receptor Ret inhibit Shh expression in dopaminergic neurons. Thus, we discovered a mechanism for neuronal subtype specific and reciprocal communication that is essential for neurochemical and structural homeostasis in the nigrostriatal circuit. These results provide integrative insights into non cell-autonomous processes likely at play in neurodegenerative conditions such as Parkinson's disease.


Subject(s)
Corpus Striatum/metabolism , Dopaminergic Neurons/metabolism , Hedgehog Proteins/metabolism , Homeostasis/physiology , Nerve Degeneration/metabolism , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Animals , Cholinergic Neurons/metabolism , Cholinergic Neurons/pathology , Corpus Striatum/pathology , Dopaminergic Neurons/pathology , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Gait/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hedgehog Proteins/genetics , Mice , Motor Activity/physiology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neural Pathways/metabolism , Neural Pathways/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Signal Transduction/physiology , Substantia Nigra/pathology
8.
J Biomed Biotechnol ; 2012: 845618, 2012.
Article in English | MEDLINE | ID: mdl-22536024

ABSTRACT

Neurological disorders can be modeled in animals so as to recreate specific pathogenic events and behavioral outcomes. Parkinson's Disease (PD) is the second most common neurodegenerative disease of an aging population, and although there have been several significant findings about the PD disease process, much of this process still remains a mystery. Breakthroughs in the last two decades using animal models have offered insights into the understanding of the PD disease process, its etiology, pathology, and molecular mechanisms. Furthermore, while cellular models have helped to identify specific events, animal models, both toxic and genetic, have replicated almost all of the hallmarks of PD and are useful for testing new neuroprotective or neurorestorative strategies. Moreover, significant advances in the modeling of additional PD features have come to light in both classic and newer models. In this review, we try to provide an updated summary of the main characteristics of these models as well as the strengths and weaknesses of what we believe to be the most popular PD animal models. These models include those produced by 6-hydroxydopamine (6-OHDA), 1-methyl-1,2,3,6-tetrahydropiridine (MPTP), rotenone, and paraquat, as well as several genetic models like those related to alpha-synuclein, PINK1, Parkin and LRRK2 alterations.


Subject(s)
Disease Models, Animal , Parkinson Disease , Animals , Haplorhini , Mice
9.
Parkinsonism Relat Disord ; 18 Suppl 1: S183-5, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22166429

ABSTRACT

Parkinson's disease (PD) is a disease of an aging population and its etiology is still unknown. In vivo models are attempts to capture as many of the hallmarks of PD as possible. To this end, a number of animal models are in use. These models parallel our thinking about the etiology of PD. Thus, herein, we discuss the most popular neurotoxin animal models, 6-hydroxydopamine and MPTP as one school of thought believes that PD is the result of a toxic insult. Since several researchers think that pesticide and herbicide use can increase the risk of developing PD, we review some of the aspects of rotenone and paraquat in rodents. Furthermore, now that we know that 10% of all PD cases are genetic in nature, we discuss some of the more common genetic rodent models of PD. None of the above models captures all of the hallmarks of PD. Thus, a given model should never be used indiscriminately to investigate every question, but should instead be carefully selected on the basis of being the most suitable model for the question being asked.


Subject(s)
Disease Models, Animal , MPTP Poisoning/physiopathology , Parkinson Disease/physiopathology , Animals , Humans , Insecticides/toxicity , MPTP Poisoning/chemically induced , MPTP Poisoning/genetics , Methamphetamine/toxicity , Oxidopamine/toxicity , Parkinson Disease/etiology , Parkinson Disease/genetics
10.
Neurotox Res ; 19(1): 63-72, 2011 Jan.
Article in English | MEDLINE | ID: mdl-19957214

ABSTRACT

In Parkinson's disease (PD), there is a progressive loss of neuromelanin (NM)-containing dopamine neurons in substantia nigra (SN) which is associated with microgliosis and presence of extracellular NM. Herein, we have investigated the interplay between microglia and human NM on the degeneration of SN dopaminergic neurons. Although NM particles are phagocytized and degraded by microglia within minutes in vitro, extracellular NM particles induce microglial activation and ensuing production of superoxide, nitric oxide, hydrogen peroxide (H2O2), and pro-inflammatory factors. Furthermore, NM produces, in a microglia-depended manner, neurodegeneration in primary ventral midbrain cultures. Neurodegeneration was effectively attenuated with microglia derived from mice deficient in macrophage antigen complex-1, a microglial integrin receptor involved in the initiation of phagocytosis. Neuronal loss was also attenuated with microglia derived from mice deficient in phagocytic oxidase, a subunit of NADPH oxidase, that is responsible for superoxide and H2O2 production, or apocynin, an NADPH oxidase inhibitor. In vivo, NM injected into rat SN produces microgliosis and a loss of tyrosine hydroxylase neurons. Thus, these results show that extracellular NM can activate microglia, which in turn may induce dopaminergic neurodegeneration in PD. Our study may have far-reaching implications, both pathogenic and therapeutic.


Subject(s)
Disease Progression , Melanins/metabolism , Microglia/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Parkinson Disease/metabolism , Animals , Dopamine , Female , Humans , Mice , Mice, Knockout , Microglia/pathology , Nerve Degeneration/pathology , Neurons/pathology , Parkinson Disease/pathology , Pregnancy , Rats , Rats, Inbred F344
11.
Prog Brain Res ; 183: 99-113, 2010.
Article in English | MEDLINE | ID: mdl-20696317

ABSTRACT

Parkinson's disease (PD) is the most common neurodegenerative movement disorder associated with a loss of dopaminergic neurons. The role of mitochondria in the aetiology of PD has been questioned for decades, mostly from the perspective of bioenergetic failure. For decades, a deficit in mitochondrial respiration was thought to be a key factor in PD neurodegeneration. However, excluding a few exceptions where a clinical picture of parkinsonism is associated with a mitochondrial DNA mutation, preclinical and clinical studies have failed to identify any genetic mutations in the genes encoding for the electron transport chain complexes in PD patients. More recently, it has been discovered that mutations in the genes encoding for Parkin, PINK1 (PTEN-induced putative kinase-1) and DJ-1 are associated with familial forms of PD and with mitochondrial alterations, including morphological abnormalities. These results have led many researchers to revisit the question of mitochondrial biology as a primary mechanism in PD pathogenesis, this time from an angle of perturbation in mitochondrial dynamics and not from the angle of a deficit in respiration.


Subject(s)
Autophagy/physiology , Cell Respiration/physiology , Mitochondria/metabolism , Parkinson Disease/pathology , Intracellular Signaling Peptides and Proteins/genetics , Mitochondria/pathology , Oncogene Proteins/genetics , Parkinson Disease/metabolism , Protein Deglycase DJ-1 , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics
12.
Ann Neurol ; 68(2): 184-92, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20695011

ABSTRACT

OBJECTIVE: Mitochondrial complex I deficits have long been associated with Parkinson disease (PD). However, it remains unknown whether such defects represent a primary event in dopaminergic neurodegeneration. METHODS: Apoptosis-inducing factor (AIF) is a mitochondrial protein that, independently of its proapoptotic properties, plays an essential physiologic role in maintaining a fully functional complex I. We used AIF-deficient harlequin (Hq) mice, which exhibit structural deficits in assembled complex I, to determine whether primary complex I defects linked to AIF depletion may cause dopaminergic neurodegeneration. RESULTS: Despite marked reductions in mitochondrial complex I protein levels, Hq mice did not display apparent alterations in the dopaminergic nigrostriatal system. However, these animals were much more susceptible to exogenous parkinsonian complex I inhibitors, such as 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Subtoxic doses of MPTP, unable to cause damage to wild-type animals, produced marked nigrostriatal dopaminergic degeneration in Hq mice. This effect was associated with exacerbated complex I inhibition and increased production of mitochondrial-derived reactive oxygen species (ROS) in Hq brain mitochondria. The antioxidant superoxide dismutase-mimetic compound tempol was able to reverse the increased susceptibility of Hq mice to MPTP. Supporting an instrumental role for mitochondrial-derived ROS in PD-related neurodegeneration, transgenic mice overexpressing mitochondrially targeted catalase exhibited an attenuation of MPTP-induced mitochondrial ROS and dopaminergic cell death. INTERPRETATION: Structural complex I alterations linked to AIF deficiency do not cause dopaminergic neurodegeneration but increase the susceptibility of dopaminergic neurons to exogenous parkinsonian neurotoxins, reinforcing the concept that genetic and environmental factors may interact in a common molecular pathway to trigger PD.


Subject(s)
Apoptosis Inducing Factor/deficiency , Dopamine/physiology , Neurons/metabolism , Neurotoxins/toxicity , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Animals , Electron Transport Complex I/deficiency , Electron Transport Complex I/genetics , Mice , Mice, Transgenic , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Neurons/drug effects , Neurons/pathology , Neurotoxins/metabolism , Parkinsonian Disorders/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism
13.
J Neurosci ; 30(3): 1166-75, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20089925

ABSTRACT

We report that rapamycin, an allosteric inhibitor of certain but not all actions of the key cellular kinase mammalian target of rapamycin (mTOR), protects neurons from death in both cellular and animal toxin models of Parkinson's disease (PD). This protective action appears to be attributable to blocked translation of RTP801/REDD1/Ddit4, a protein that is induced in cell and animal models of PD and in affected neurons of PD patients and that causes neuron death by leading to dephosphorylation of the survival kinase Akt. In support of this mechanism, in PD models, rapamycin spares phosphorylation of Akt at a site critical for maintenance of its survival-promoting activity. The capacity of rapamycin to provide neuroprotection in PD models appears to arise from its selective suppression of some but not all actions of mTOR, as indicated by the contrasting finding that Torin1, a full catalytic mTOR inhibitor, is not protective and induces Akt dephosphorylation and neuron death.


Subject(s)
Neurons/drug effects , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/pathology , Parkinsonian Disorders/prevention & control , Sirolimus/therapeutic use , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Analysis of Variance , Animals , Cell Death/drug effects , Cycloheximide/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factor/pharmacology , Oxidopamine/toxicity , PC12 Cells , Parkinson Disease , Parkinsonian Disorders/chemically induced , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Serine/metabolism , TOR Serine-Threonine Kinases , Time Factors , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/pharmacology , Transfection/methods , Tyrosine 3-Monooxygenase/metabolism
14.
Free Radic Biol Med ; 47(7): 1049-56, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19616617

ABSTRACT

DNA damage is a proposed pathogenic factor in neurodegenerative disorders such as Parkinson disease. To probe the underpinning mechanism of such neuronal perturbation, we sought to produce an experimental model of DNA damage. We thus first assessed DNA damage by in situ nick translation and emulsion autoradiography in the mouse brain after administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP; 4 x 20 mg/kg, ip, every 2 h), a neurotoxin known to produce a model of Parkinson disease. Here we show that DNA strand breaks occur in vivo in this mouse model of Parkinson disease with kinetics and a topography that parallel the degeneration of substantia nigra neurons, as assessed by FluoroJade labeling. Previously, nitric oxide synthase and cyclooxygenase-2 (Cox-2) were found to modulate MPTP-induced dopaminergic neuronal death. We thus assessed the contribution of these enzymes to DNA damage in mice lacking neuronal nitric oxide synthase (nNOS), inducible nitric oxide synthase (iNOS), or Cox-2. We found that the lack of Cox-2 and nNOS activities but not of iNOS activity attenuated MPTP-related DNA damage. We also found that not only nuclear, but also mitochondrial, DNA is a target for the MPTP insult. These results suggest that the loss of genomic integrity can be triggered by the concerted actions of nNOS and Cox-2 and provide further support to the view that DNA damage may contribute to the neurodegenerative process in Parkinson disease.


Subject(s)
Cyclooxygenase 2/metabolism , Disease Models, Animal , Nitric Oxide Synthase Type I/metabolism , Parkinsonian Disorders/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cyclooxygenase 2/deficiency , DNA Damage , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type I/deficiency , Oxidative Stress/drug effects , Parkinsonian Disorders/enzymology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Time Factors
15.
Nat Neurosci ; 12(7): 826-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19503083

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic cause of Parkinson's disease. We created a LRRK2 transgenic mouse model that recapitulates cardinal features of the disease: an age-dependent and levodopa-responsive slowness of movement associated with diminished dopamine release and axonal pathology of nigrostriatal dopaminergic projection. These mice provide a valid model of Parkinson's disease and are a resource for the investigation of pathogenesis and therapeutics.


Subject(s)
Disease Models, Animal , Mice, Transgenic , Parkinson Disease , Protein Serine-Threonine Kinases/genetics , Age Factors , Animals , Antiparkinson Agents/therapeutic use , Apomorphine/therapeutic use , Brain/drug effects , Brain/pathology , Brain/physiopathology , Chromosomes, Artificial, Bacterial , Dopamine/metabolism , Dopamine Uptake Inhibitors/pharmacology , Gene Transfer Techniques , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Levodopa/therapeutic use , Mice , Movement Disorders/drug therapy , Movement Disorders/genetics , Mutation, Missense , Neurons/pathology , Neurons/physiology , Nomifensine/pharmacology , Parkinson Disease/genetics , Parkinson Disease/pathology , Parkinson Disease/physiopathology , Protein Serine-Threonine Kinases/metabolism
16.
Proc Natl Acad Sci U S A ; 106(19): 8043-8, 2009 May 12.
Article in English | MEDLINE | ID: mdl-19416912

ABSTRACT

Toxic organic cations can damage nigrostriatal dopaminergic pathways as seen in most parkinsonian syndromes and in some cases of illicit drug exposure. Here, we show that the organic cation transporter 3 (Oct3) is expressed in nondopaminergic cells adjacent to both the soma and terminals of midbrain dopaminergic neurons. We hypothesized that Oct3 contributes to the dopaminergic damage by bidirectionally regulating the local bioavailability of toxic species. Consistent with this view, Oct3 deletion and pharmacological inhibition hampers the release of the toxic organic cation 1-methyl-4-phenylpyridinium from astrocytes and protects against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration in mice. Furthermore, Oct3 deletion impairs the removal of the excess extracellular dopamine induced by methamphetamine and enhances striatal dopaminergic terminal damage caused by this psychostimulant. These results may have far-reaching implications for our understanding of the mechanism of cell death in a wide range of neurodegenerative diseases and may open new avenues for neuroprotective intervention.


Subject(s)
Dopamine/metabolism , Neurodegenerative Diseases/metabolism , Organic Cation Transport Proteins/physiology , Substantia Nigra/metabolism , Animals , Astrocytes/metabolism , Corpus Striatum/metabolism , Glutamic Acid/metabolism , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Neurons/metabolism , Organic Cation Transport Proteins/metabolism , Time Factors
17.
PLoS One ; 3(7): e2740, 2008 Jul 23.
Article in English | MEDLINE | ID: mdl-18648532

ABSTRACT

BACKGROUND: Innate neuroimmune dysfunction is a pathobiological feature of amyotrophic lateral sclerosis (ALS). However, links, if any, between disease and adaptive immunity are poorly understood. Thus, the role of T cell immunity in disease was investigated in human G93A superoxide dismutase 1 (SOD1) transgenic (Tg) mice and subsequently in ALS patients. METHODS AND FINDINGS: Quantitative and qualitative immune deficits in lymphoid cell and T cell function were seen in G93A-SOD1 Tg mice. Spleens of Tg animals showed reductions in size, weight, lymphocyte numbers, and morphological deficits at terminal stages of disease compared to their wild-type (Wt) littermates. Spleen sizes and weights of pre-symptomatic Tg mice were unchanged, but deficits were readily seen in T cell proliferation coincident with increased annexin-V associated apoptosis and necrosis of lymphocytes. These lymphoid deficits paralleled failure of Copolymer-1 (COP-1) immunization to affect longevity. In addition, among CD4(+) T cells in ALS patients, levels of CD45RA(+) (naïve) T cells were diminished, while CD45RO(+) (memory) T cells were increased compared to age-matched caregivers. In attempts to correct mutant SOD1 associated immune deficits, we reconstituted SOD1 Tg mice with unfractionated naïve lymphocytes or anti-CD3 activated CD4(+)CD25(+) T regulatory cells (Treg) or CD4(+)CD25(-) T effector cells (Teff) from Wt donor mice. While naive lymphocytes failed to enhance survival, both polyclonal-activated Treg and Teff subsets delayed loss of motor function and extended survival; however, only Treg delayed neurological symptom onset, whereas Teff increased latency between disease onset and entry into late stage. CONCLUSIONS: A profound and progressive immunodeficiency is operative in G93A-SOD1 mice and is linked to T cell dysfunction and the failure to elicit COP-1 neuroprotective immune responses. In preliminary studies T cell deficits were also observed in human ALS. These findings, taken together, suggest caution in ascribing vaccination outcomes when these animal models of human ALS are used for study. Nonetheless, the abilities to improve neurological function and life expectancy in G93A-SOD1 Tg mice by reconstitution with activated T cells do provide opportunities for therapeutic intervention.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/immunology , Superoxide Dismutase/genetics , Animals , Apoptosis , CD4-Positive T-Lymphocytes/cytology , Humans , Immune System , Interleukin-2 Receptor alpha Subunit/biosynthesis , Lymphocytes/cytology , Mice , Mice, Transgenic , Necrosis , Phenotype , Spleen/metabolism , T-Lymphocytes/immunology
18.
Proc Natl Acad Sci U S A ; 104(19): 8161-6, 2007 May 08.
Article in English | MEDLINE | ID: mdl-17483459

ABSTRACT

Dysfunction of mitochondrial complex I is associated with a wide spectrum of neurodegenerative disorders, including Parkinson's disease (PD). In rodents, inhibition of complex I leads to degeneration of dopaminergic neurons of the substantia nigra pars compacta (SNpc), as seen in PD, through activation of mitochondria-dependent apoptotic molecular pathways. In this scenario, complex I blockade increases the soluble pool of cytochrome c in the mitochondrial intermembrane space through oxidative mechanisms, whereas activation of pro-cell death protein Bax is actually necessary to trigger neuronal death by permeabilizing the outer mitochondrial membrane and releasing cytochrome c into the cytosol. Activation of Bax after complex I inhibition relies on its transcriptional induction and translocation to the mitochondria. How complex I deficiency leads to Bax activation is currently unknown. Using gene-targeted mice, we show that the tumor suppressor p53 mediates Bax transcriptional induction after PD-related complex I blockade in vivo, but it does not participate in Bax mitochondrial translocation in this model, either by a transcription-independent mechanism or through the induction of BH3-only proteins Puma or Noxa. Instead, Bax mitochondrial translocation in this model relies mainly on the JNK-dependent activation of the BH3-only protein Bim. Targeting either Bax transcriptional induction or Bax mitochondrial translocation results in a marked attenuation of SNpc dopaminergic cell death caused by complex I inhibition. These results provide further insight into the pathogenesis of PD neurodegeneration and identify molecular targets of potential therapeutic significance for this disabling neurological illness.


Subject(s)
Mitochondria/physiology , Neurodegenerative Diseases/etiology , Parkinsonian Disorders/pathology , Substantia Nigra/pathology , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , DNA Damage , Electron Transport Complex I/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Protein Transport , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/physiology , bcl-2-Associated X Protein/metabolism
19.
Nat Protoc ; 2(1): 141-51, 2007.
Article in English | MEDLINE | ID: mdl-17401348

ABSTRACT

This protocol describes our method of producing a reliable mouse model of Parkinson's disease (PD) using the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We discuss the particulars of the model, provide key references and outline what investigators need to know to develop the MPTP mouse model of PD safely and successfully. Completion of this protocol depends on the regimen of MPTP used and on the actual planned studies, which often range from 7 to 30 d. This protocol calls for implementation of safety measures and for the acquisition of several pieces of equipment, which are a one-time investment worth making if one elects to use this model on a regular basis.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Disease Models, Animal , Parkinsonian Disorders/chemically induced , 3,4-Dihydroxyphenylacetic Acid , Animals , Dopamine/metabolism , Mice , Substantia Nigra/cytology , Tyrosine 3-Monooxygenase
20.
Eur J Neurosci ; 24(7): 1874-84, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17067291

ABSTRACT

The aetiology of idiopathic Parkinson's disease (PD) is poorly defined but environmental aggression may be relevant. Here, we report a new model of PD in mice, based on chronic inoculation with neurotoxins in the nasal cavity, which is a natural route of contact with the environment. C57BL/6 mice, submitted to daily intranasal inoculation with MPTP for 30 days, developed motor deficits that correlated with a progressive and severe depletion of striatal dopamine levels, and loss of tyrosine hydroxylase and dopamine transporter staining in substantia nigra and striatum. Moreover, mice intranasally inoculated with MPTP developed strong astrogliosis and microgliosis in substantia nigra and striatum. Consistent with these observations, a role for oxidant aggression was demonstrated by increased levels of Mn-superoxide dismutase. However, alpha-synuclein aggregation was not observed. This new animal model provides a new tool for studying PD symptoms that develop slowly over time, and it may be used to asses risk from environmental neurotoxins.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/administration & dosage , Administration, Intranasal , Disease Models, Animal , Neurotoxins/administration & dosage , Parkinson Disease , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Acetylcholinesterase/metabolism , Animals , Behavior, Animal , Blotting, Western/methods , Brain/metabolism , Brain/pathology , Dopamine/metabolism , Dose-Response Relationship, Drug , Drug Administration Routes , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurotoxins/metabolism , Parkinson Disease/etiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Synucleins/genetics , Synucleins/metabolism , Time Factors , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...