Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Front Aging Neurosci ; 14: 1064057, 2022.
Article in English | MEDLINE | ID: mdl-36533178

ABSTRACT

Despite many pharmacological and surgical treatments addressing the symptoms of Parkinson's disease, there are no approved treatments that slow disease progression. Genetic discoveries in the last 20 years have increased our understanding of the molecular contributors to Parkinson's pathophysiology, uncovered many druggable targets and pathways, and increased investment in treatments that might slow or stop the disease process. Longitudinal, observational studies are dissecting Parkinson's disease heterogeneity and illuminating the importance of molecularly defined subtypes more likely to respond to targeted interventions. Indeed, clinical and pathological differences seen within and across carriers of PD-associated gene mutations suggest the existence of greater biological complexity than previously appreciated and increase the likelihood that targeted interventions based on molecular characteristics will be beneficial. This article offers our current perspective on the promise and current challenges in subtype identification and precision medicine approaches in Parkinson's disease.

2.
Sci Transl Med ; 12(540)2020 04 22.
Article in English | MEDLINE | ID: mdl-32321864

ABSTRACT

The kinase-activating mutation G2019S in leucine-rich repeat kinase 2 (LRRK2) is one of the most common genetic causes of Parkinson's disease (PD) and has spurred development of LRRK2 inhibitors. Preclinical studies have raised concerns about the safety of LRRK2 inhibitors due to histopathological changes in the lungs of nonhuman primates treated with two of these compounds. Here, we investigated whether these lung effects represented on-target pharmacology and whether they were reversible after drug withdrawal in macaques. We also examined whether treatment was associated with pulmonary function deficits. We conducted a 2-week repeat-dose toxicology study in macaques comparing three different LRRK2 inhibitors: GNE-7915 (30 mg/kg, twice daily as a positive control), MLi-2 (15 and 50 mg/kg, once daily), and PFE-360 (3 and 6 mg/kg, once daily). Subsets of animals dosed with GNE-7915 or MLi-2 were evaluated 2 weeks after drug withdrawal for lung function. All compounds induced mild cytoplasmic vacuolation of type II lung pneumocytes without signs of lung degeneration, implicating on-target pharmacology. At low doses of PFE-360 or MLi-2, there was ~50 or 100% LRRK2 inhibition in brain tissue, respectively, but histopathological lung changes were either absent or minimal. The lung effect was reversible after dosing ceased. Lung function tests demonstrated that the histological changes in lung tissue induced by MLi-2 and GNE-7915 did not result in pulmonary deficits. Our results suggest that the observed lung effects in nonhuman primates in response to LRRK2 inhibitors should not preclude clinical testing of these compounds for PD.


Subject(s)
Parkinson Disease , Animals , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Lung , Morpholines , Mutation , Primates , Pyrimidines , Pyrroles
3.
J Parkinsons Dis ; 6(4): 703-707, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27497486

ABSTRACT

Recent research suggests that in Parkinson's disease the long, thin and unmyelinated axons of dopaminergic neurons degenerate early in the disease process. We organized a workshop entitled 'Axonal Pathology in Parkinson's disease', on March 23rd, 2016, in Cleveland, Ohio with the goals of summarizing the state-of-the-art and defining key gaps in knowledge. A group of eight research leaders discussed new developments in clinical pathology, functional imaging, animal models, and mechanisms of degeneration including neuroinflammation, autophagy and axonal transport deficits. While the workshop focused on PD, comparisons were made to other neurological conditions where axonal degeneration is well recognized.


Subject(s)
Axons/pathology , Congresses as Topic , Dopaminergic Neurons/pathology , Parkinson Disease/pathology , Animals , Humans
4.
J Parkinsons Dis ; 6(3): 519-22, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27434298

ABSTRACT

We discuss a report in the current issue on clinical and biochemical findings from a safety trial using the cAbl tyrosine kinase inhibitor Nilotinib (150 mg or 300 mg given daily for 24 weeks) in a small group of patients with either advanced Parkinson's disease or Dementia with Lewy Bodies. Despite some side effects (one serious), the authors claim that Nilotinib, which is normally used at much higher doses for treating leukemia, is safe and tolerated. Furthermore, they report a possible benefit on motor and cognitive outcomes. We debate the safety of Nilotinib and the reported efficacy signals. We emphasize that due to the small sample size, and lack of a control group, it is impossible to rule out a placebo effect. We briefly discuss a range of aspects surrounding the current and possible future use of this cAbl inhibitor in patients with alpha-synucleinopathy, and what must now be done to obtain definitive information about its safety and efficacy in this population of patients.


Subject(s)
Dementia , Lewy Body Disease , Parkinson Disease , Humans , Pyrimidines
5.
6.
Neurobiol Dis ; 70: 190-203, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24969022

ABSTRACT

Recessively inherited loss-of-function mutations in the PTEN-induced putative kinase 1(Pink1), DJ-1 (Park7) and Parkin (Park2) genes are linked to familial cases of early-onset Parkinson's disease (PD). As part of its strategy to provide more tools for the research community, The Michael J. Fox Foundation for Parkinson's Research (MJFF) funded the generation of novel rat models with targeted disruption ofPink1, DJ-1 or Parkin genes and determined if the loss of these proteins would result in a progressive PD-like phenotype. Pathological, neurochemical and behavioral outcome measures were collected at 4, 6 and 8months of age in homozygous KO rats and compared to wild-type (WT) rats. Both Pink1 and DJ-1 KO rats showed progressive nigral neurodegeneration with about 50% dopaminergic cell loss observed at 8 months of age. ThePink1 KO and DJ-1 KO rats also showed a two to three fold increase in striatal dopamine and serotonin content at 8 months of age. Both Pink1 KO and DJ-1 KO rats exhibited significant motor deficits starting at 4months of age. However, Parkin KO rats displayed normal behaviors with no neurochemical or pathological changes. These results demonstrate that inactivation of the Pink1 or DJ-1 genes in the rat produces progressive neurodegeneration and early behavioral deficits, suggesting that these recessive genes may be essential for the survival of dopaminergic neurons in the substantia nigra (SN). These MJFF-generated novel rat models will assist the research community to elucidate the mechanisms by which these recessive genes produce PD pathology and potentially aid in therapeutic development.


Subject(s)
Microtubule-Associated Proteins/deficiency , Parkinsonian Disorders/physiopathology , Phenotype , Protein Kinases/deficiency , Ubiquitin-Protein Ligases/deficiency , Aging , Animals , Animals, Genetically Modified , Brain/pathology , Brain/physiopathology , Dopamine/metabolism , Dopaminergic Neurons/pathology , Dopaminergic Neurons/physiology , Gene Knockout Techniques , Genes, Recessive , Male , Microtubule-Associated Proteins/genetics , Motor Activity/physiology , Parkinsonian Disorders/genetics , Parkinsonian Disorders/pathology , Protein Deglycase DJ-1 , Protein Kinases/genetics , Rats, Long-Evans , Serotonin/metabolism , Ubiquitin-Protein Ligases/genetics
7.
Mov Disord ; 29(6): 772-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24610195

ABSTRACT

Amantadine, an N-methyl-D-aspartate glutamate receptor antagonist, is currently the only pharmacological treatment for levodopa-induced dyskinesia (LID) in Parkinson's disease (PD), but causes adverse effects on the central nervous system at therapeutic doses. Fenobam, a negative modulator of metabotropic glutamate receptor subtype 5, has recently been reported to attenuate LID in MPTP-treated macaques. The aim of the current study was to investigate the treatment interactions of fenobam and amantadine on LID in the MPTP-treated macaque model of PD. The antidyskinetic and -parkinsonian effects were measured after administration of fenobam (10-30 mg/kg) and amantadine (10-30 mg/kg) alone and in combination. Fenobam (30 mg/kg) and amantadine (30 mg/kg) alone reduced LID, whereas lower doses of either drug did not cause any significant effects. A combined treatment of fenobam and amantadine at subthreshold doses (10 and 20 mg/kg) significantly reduced LID without worsening PD disability. These data suggest that a low-dose combination of fenobam and amantadine can be used for alleviating dyskinesia without causing adverse motor effects. Such combined therapies may offer a new therapeutic strategy for treatment of LID in PD patients.


Subject(s)
Amantadine/therapeutic use , Dyskinesia, Drug-Induced/drug therapy , Excitatory Amino Acid Antagonists/therapeutic use , Imidazoles/therapeutic use , Analysis of Variance , Animals , Antiparkinson Agents/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , Dyskinesia, Drug-Induced/etiology , Female , Levodopa/adverse effects , MPTP Poisoning/drug therapy , Macaca fascicularis
8.
PLoS One ; 8(11): e80705, 2013.
Article in English | MEDLINE | ID: mdl-24244710

ABSTRACT

The objective of this study was to evaluate the pathology time course of the LRRK2 knockout rat model of Parkinson's disease at 1-, 2-, 4-, 8-, 12-, and 16-months of age. The evaluation consisted of histopathology and ultrastructure examination of selected organs, including the kidneys, lungs, spleen, heart, and liver, as well as hematology, serum, and urine analysis. The LRRK2 knockout rat, starting at 2-months of age, displayed abnormal kidney staining patterns and/or morphologic changes that were associated with higher serum phosphorous, creatinine, cholesterol, and sorbitol dehydrogenase, and lower serum sodium and chloride compared to the LRRK2 wild-type rat. Urinalysis indicated pronounced changes in LRRK2 knockout rats in urine specific gravity, total volume, urine potassium, creatinine, sodium, and chloride that started as early as 1- to 2-months of age. Electron microscopy of 16-month old LRRK2 knockout rats displayed an abnormal kidney, lung, and liver phenotype. In contrast, there were equivocal or no differences in the heart and spleen of LRRK2 wild-type and knockout rats. These findings partially replicate data from a recent study in 4-month old LRRK2 knockout rats and expand the analysis to demonstrate that the renal and possibly lung and liver abnormalities progress with age. The characterization of LRRK2 knockout rats may prove to be extremely valuable in understanding potential safety liabilities of LRRK2 kinase inhibitor therapeutics for treating Parkinson's disease.


Subject(s)
Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Animals , Kidney/metabolism , Kidney/pathology , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Liver/metabolism , Liver/pathology , Lung/metabolism , Lung/pathology , Male , Phenotype , Rats , Rats, Mutant Strains , Spleen/metabolism , Spleen/pathology
9.
Dis Model Mech ; 6(6): 1316-24, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24046356

ABSTRACT

Progress in Parkinson's disease (PD) research and therapeutic development is hindered by many challenges, including a need for robust preclinical animal models. Limited availability of these tools is due to technical hurdles, patent issues, licensing restrictions and the high costs associated with generating and distributing these animal models. Furthermore, the lack of standardization of phenotypic characterization and use of varying methodologies has made it difficult to compare outcome measures across laboratories. In response, The Michael J. Fox Foundation for Parkinson's Research (MJFF) is directly sponsoring the generation, characterization and distribution of preclinical rodent models, enabling increased access to these crucial tools in order to accelerate PD research. To date, MJFF has initiated and funded the generation of 30 different models, which include transgenic or knockout models of PD-relevant genes such as Park1 (also known as Park4 and SNCA), Park8 (LRRK2), Park7 (DJ-1), Park6 (PINK1), Park2 (Parkin), VPS35, EiF4G1 and GBA. The phenotypic characterization of these animals is performed in a uniform and streamlined manner at independent contract research organizations. Finally, MJFF created a central repository at The Jackson Laboratory (JAX) that houses both non-MJFF and MJFF-generated preclinical animal models. Funding from MJFF, which subsidizes the costs involved in transfer, rederivation and colony expansion, has directly resulted in over 2500 rodents being distributed to the PD community for research use.


Subject(s)
Biomedical Research , Models, Animal , Parkinson Disease , Animals , Animals, Genetically Modified , Humans , Parkinson Disease/genetics , Promoter Regions, Genetic
10.
Nat Med ; 19(2): 127, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23389601
11.
Mov Disord ; 27(13): 1606-11, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23115047

ABSTRACT

Improved symptomatic and disease-modifying treatments are needed for Parkinson's disease (PD). Although significant advances have been made in the understanding of PD etiology, the translation of these discoveries into novel transformative therapies has been limited as a result of systemic challenges in PD drug development. Preclinical testing lacks clear standards and prioritization criteria for advancing therapies to the clinic. Clinical testing is marked by expensive, long, and uninformative studies. In parallel to these scientific challenges, funding of late-stage drug development has become increasingly scarce and risk averse. In this context, novel models of collaboration and funding are opening up new avenues for pursuing treatments. This review will discuss the most critical challenges in PD drug development and the innovative approaches being developed to overcome these hurdles.


Subject(s)
Clinical Trials as Topic , Parkinson Disease/physiopathology , Parkinson Disease/therapy , Clinical Trials as Topic/economics , Humans , Parkinson Disease/economics
12.
Sci Transl Med ; 3(79): 79ps14, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21508309

ABSTRACT

Biomarkers for detecting the early stages of Parkinson's disease (PD) could accelerate development of new treatments. Such biomarkers could be used to identify individuals at risk for developing PD, to improve early diagnosis, to track disease progression with precision, and to test the efficacy of new treatments. Although some progress has been made, there are many challenges associated with developing biomarkers for detecting PD in its earliest stages.


Subject(s)
Biomarkers/metabolism , Disease Progression , Parkinson Disease/diagnosis , Parkinson Disease/physiopathology , Clinical Trials as Topic , Diagnostic Imaging , Humans , Parkinson Disease/pathology
14.
J Neurochem ; 100(6): 1469-79, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17241123

ABSTRACT

Parkinson's disease (PD) has been linked to mitochondrial dysfunction and pesticide exposure. The pesticide rotenone (ROT) inhibits complex I and reproduces features of PD in animal models, suggesting that environmental agents that inhibit complex I may contribute to PD. We have previously demonstrated that ROT toxicity is dependent upon complex I inhibition and that oxidative stress is the primary mechanism of toxicity. In this study, we examined the in vitro toxicity and mechanism of action of several putative complex I inhibitors that are commonly used as pesticides. The rank order of toxicity of pesticides to neuroblastoma cells was pyridaben > rotenone > fenpyroximate > fenazaquin > tebunfenpyrad. A similar order of potency was observed for reduction of ATP levels and competition for (3)H-dihydrorotenone (DHR) binding to complex I, with the exception of pyridaben (PYR). Neuroblastoma cells stably expressing the ROT-insensitive NADH dehydrogenase of Saccharomyces cerevisiae (NDI1) were resistant to these pesticides, demonstrating the requirement of complex I inhibition for toxicity. We further found that PYR was a more potent inhibitor of mitochondrial respiration and caused more oxidative damage than ROT. The oxidative damage could be attenuated by NDI1 or by the antioxidants alpha-tocopherol and coenzyme Q(10). PYR was also highly toxic to midbrain organotypic slices. These data demonstrate that, in addition to ROT, several commercially used pesticides directly inhibit complex I, cause oxidative damage, and suggest that further study is warranted into environmental agents that inhibit complex I for their potential role in PD.


Subject(s)
Adenosine Triphosphate/metabolism , Electron Transport Complex I/metabolism , Mitochondria/drug effects , Pesticides/toxicity , Animals , Animals, Newborn , Antioxidants/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Interactions , Humans , In Vitro Techniques , Male , Mesencephalon/ultrastructure , NADH Dehydrogenase/pharmacology , Neuroblastoma , Pesticides/chemistry , Protein Carbonylation/drug effects , Rats , Rats, Inbred Lew , Rotenone/analogs & derivatives , Rotenone/pharmacokinetics , Saccharomyces cerevisiae Proteins/pharmacology , Tyrosine 3-Monooxygenase/metabolism , alpha-Tocopherol/pharmacology
15.
Toxicol Sci ; 95(1): 196-204, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17038483

ABSTRACT

Administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to mice and nonhuman primates causes a parkinsonian disorder characterized by a loss of dopamine-producing neurons in the substantia nigra and corresponding motor deficits. MPTP has been proposed to exert its neurotoxic effects through a variety of mechanisms, including inhibition of complex I of the mitochondrial respiratory chain, displacement of dopamine from vesicular stores, and formation of reactive oxygen species from mitochondrial or cytosolic sources. However, the mechanism of MPTP-induced neurotoxicity is still a matter of debate. Recently, we reported that the yeast single-subunit nicotinamide adenine dinucleotide (reduced) dehydrogenase (NDI1) is resistant to rotenone, a complex I inhibitor that produces a parkinsonian syndrome in rats, and that overexpression of NDI1 in SK-N-MC cells prevents the toxicity of rotenone. In this study, we used viral-mediated overexpression of NDI1 in SK-N-MC cells and animals to determine the relative contribution of complex I inhibition in the toxicity of MPTP. In cell culture, NDI1 overexpression abolished the toxicity of 1-methyl-4-phenylpyridinium, the active metabolite of MPTP. Overexpression of NDI1 through stereotactic administration of a viral vector harboring the NDI1 gene into the substantia nigra protected mice from both the neurochemical and behavioral deficits elicited by MPTP. These data identify inhibition of complex I as a requirement for dopaminergic neurodegeneration and subsequent behavioral deficits produced by MPTP. Furthermore, combined with reports of a complex I defect in Parkinson's disease (PD) patients, the present study affirms the utility of MPTP in understanding the molecular mechanisms underlying dopaminergic neurodegeneration in PD.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Dopamine/metabolism , Electron Transport Complex I/antagonists & inhibitors , MPTP Poisoning/metabolism , Motor Skills Disorders/metabolism , NADH Dehydrogenase/biosynthesis , Saccharomyces cerevisiae Proteins/biosynthesis , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Animals , Behavior, Animal , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Death/drug effects , Cell Line, Tumor , Dependovirus/genetics , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Electron Transport Complex I/metabolism , Genetic Therapy , Genetic Vectors , Humans , MPTP Poisoning/chemically induced , MPTP Poisoning/pathology , MPTP Poisoning/prevention & control , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/metabolism , Motor Activity/drug effects , Motor Skills Disorders/chemically induced , Motor Skills Disorders/pathology , Motor Skills Disorders/prevention & control , NADH Dehydrogenase/genetics , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transfection
16.
Mov Disord ; 21(2): 136-41, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16470786

ABSTRACT

The development of a neuroprotective or neuroregenerative therapy for Parkinson's disease (PD) would be a major therapeutic advance. Unfortunately, results from a recent controlled clinical study delivering the neurotrophic factor, glial-derived neurotrophic factor (GDNF), directly into brain did not demonstrate efficacy and safety of such a treatment. A critical review of available data suggests that there are questions that need to be answered before the future of GDNF as a therapy for PD can be determined.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factors/administration & dosage , Nerve Regeneration/drug effects , Neuroprotective Agents/administration & dosage , Parkinson Disease/drug therapy , Parkinsonian Disorders/drug therapy , Putamen/drug effects , Animals , Catheters, Indwelling , Clinical Trials as Topic , Dose-Response Relationship, Drug , Glial Cell Line-Derived Neurotrophic Factors/adverse effects , Humans , Magnetic Resonance Imaging , Multicenter Studies as Topic , Neuroprotective Agents/adverse effects , Putamen/pathology
17.
Neurobiol Dis ; 22(2): 404-20, 2006 May.
Article in English | MEDLINE | ID: mdl-16439141

ABSTRACT

Sporadic Parkinson's disease (PD) is most likely caused by a combination of environmental exposures and genetic susceptibilities, although there are rare monogenic forms of the disease. Mitochondrial impairment at complex I, oxidative stress, alpha-synuclein aggregation, and dysfunctional protein degradation, have been implicated in PD pathogenesis, but how they are related to each other is unclear. To further evaluated PD pathogenesis here, we used in vivo and in vitro models of chronic low-grade complex I inhibition with the pesticide rotenone. Chronic rotenone exposure in vivo caused oxidative modification of DJ-1, accumulation of alpha-synuclein, and proteasomal impairment. Interestingly, the effects become more regionally restricted such that systemic complex I inhibition eventually results in highly selective degeneration of the nigrostriatal pathway. DJ-1 modifications, alpha-synuclein accumulation, and proteasomal dysfunction were also seen in vitro and these effects could be prevented with alpha-tocopherol. Thus, chronic exposure to a pesticide and mitochondrial toxin brings into play three systems, DJ-1, alpha-synuclein, and the ubiquitin-proteasome system, and implies that mitochondrial dysfunction and oxidative stress link environmental and genetic forms of the disease.


Subject(s)
Nerve Degeneration/chemically induced , Oncogene Proteins/drug effects , Parkinsonian Disorders/chemically induced , Proteasome Endopeptidase Complex/drug effects , Rotenone/toxicity , Ubiquitin/drug effects , alpha-Synuclein/drug effects , Animals , Cell Line, Tumor , Disease Models, Animal , Electron Transport Complex I/drug effects , Electron Transport Complex I/physiology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Humans , Insecticides/toxicity , Male , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neurons/drug effects , Neurons/metabolism , Oncogene Proteins/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/physiopathology , Peroxiredoxins , Proteasome Endopeptidase Complex/metabolism , Protein Deglycase DJ-1 , Rats , Rats, Inbred Lew , Signal Transduction/drug effects , Signal Transduction/physiology , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/physiopathology , Ubiquitin/metabolism , alpha-Synuclein/metabolism
18.
Toxicol Sci ; 88(1): 193-201, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16141438

ABSTRACT

Paraquat, MPTP, and rotenone reproduce features of Parkinson's disease (PD) in experimental animals. The exact mechanisms by which these compounds damage the dopamine system are not firmly established, but selective damage to dopamine neurons and inhibition of complex I are thought to be involved. We and others have previously documented that the toxic metabolite of MPTP, MPP+, is transported into dopamine neurons through the dopamine transporter (DAT), while rotenone is not transported by DAT. We have also demonstrated the requirement for complex I inhibition and oxidative damage in the dopaminergic neurodegeneration produced by rotenone. Based on structural similarity to MPP+, it has been proposed that paraquat exerts selective dopaminergic toxicity through transport by the DAT and subsequent inhibition of mitochondrial complex I. In this study we report that paraquat is neither a substrate nor inhibitor of DAT. We also demonstrate that in vivo exposure to MPTP and rotenone, but not paraquat, inhibits binding of 3H-dihydrorotenone to complex I in brain mitochondria. Rotenone and MPP+ were both effective inhibitors of complex I activity in isolated brain mitochondria, while paraquat exhibited weak inhibitory effects only at millimolar concentrations. These data indicate that, despite the apparent structural similarity to MPP+, paraquat exerts its deleterious effects on dopamine neurons in a manner that is unique from rotenone and MPTP.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Dopamine Agents/toxicity , Herbicides/toxicity , Insecticides/toxicity , Neurons/drug effects , Paraquat/toxicity , Rotenone/toxicity , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/metabolism , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Dopamine Agents/metabolism , Dose-Response Relationship, Drug , Herbicides/metabolism , Humans , Insecticides/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Neuroblastoma , Neurons/metabolism , Neurons/pathology , Paraquat/metabolism , Rotenone/metabolism , Substrate Specificity/drug effects
20.
Brain Res Mol Brain Res ; 134(1): 109-18, 2005 Mar 24.
Article in English | MEDLINE | ID: mdl-15790535

ABSTRACT

Rotenone, a pesticide and complex I inhibitor, causes nigrostriatal degeneration similar to Parkinson disease pathology in a chronic, systemic, in vivo rodent model [M. Alam, W.J. Schmidt, Rotenone destroys dopaminergic neurons and induces parkinsonian symptoms in rats, Behav. Brain Res. 136 (2002) 317-324; R. Betarbet, T.B. Sherer, G. MacKenzie, M. Garcia-Osuna, A.V. Panov, J.T. Greenamyre, Chronic systemic pesticide exposure reproduces features of Parkinson's disease, Nat. Neurosci. 3 (2000) 1301-1306; S.M. Fleming, C. Zhu, P.O. Fernagut, A. Mehta, C.D. DiCarlo, R.L. Seaman, M.F. Chesselet, Behavioral and immunohistochemical effects of chronic intravenous and subcutaneous infusions of varying doses of rotenone, Exp. Neurol. 187 (2004) 418-429; T.B. Sherer, J.H. Kim, R. Betarbet, J.T. Greenamyre, Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation, Exp. Neurol. 179 (2003) 9-16.]. To better investigate the role of mitochondria and complex I inhibition in chronic, progressive neurodegenerative disease, we developed methods for long-term culture of rodent postnatal midbrain organotypic slices. Chronic complex I inhibition over weeks by low dose (10-50 nM) rotenone in this system lead to dose- and time-dependent destruction of substantia nigra pars compacta neuron processes, morphologic changes, some neuronal loss, and decreased tyrosine hydroxylase (TH) protein levels. Chronic complex I inhibition also caused oxidative damage to proteins, measured by protein carbonyl levels. This oxidative damage was blocked by the antioxidant alpha-tocopherol (vitamin E). At the same time, alpha-tocopherol also blocked rotenone-induced reductions in TH protein and TH immunohistochemical changes. Thus, oxidative damage is a primary mechanism of mitochondrial toxicity in intact dopaminergic neurons. The organotypic culture system allows close study of this and other interacting mechanisms over a prolonged time period in mature dopaminergic neurons with intact processes, surrounding glia, and synaptic connections.


Subject(s)
Dopamine/metabolism , Nerve Degeneration/chemically induced , Neurons/drug effects , Oxidative Stress/drug effects , Rotenone/pharmacology , Substantia Nigra/cytology , Animals , Animals, Newborn , Antioxidants/pharmacology , Blotting, Western/methods , Cell Count/methods , Dose-Response Relationship, Drug , Drug Interactions , Gene Expression Regulation/drug effects , Immunohistochemistry/methods , Nerve Degeneration/prevention & control , Neurons/metabolism , Organ Culture Techniques , Oxidative Stress/physiology , Rats , Rotenone/adverse effects , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Time Factors , Tyrosine 3-Monooxygenase/metabolism , alpha-Tocopherol/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL