Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
IBRO Neurosci Rep ; 12: 217-227, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35321527

ABSTRACT

Exercise and consumption of plant-based foods rich in polyphenols are attractive therapeutic approaches for the prevention and treatment of Parkinson's disease (PD). Few studies, however, have examined the neuroprotective efficacy of combining these treatment modalities against PD. Therefore we investigated whether combining voluntary running and consumption of blueberry juice (BBJ) was more efficacious against 6-hydroxydopamine (6-OHDA) toxicity than either treatment alone. Four weeks of running before and after intrastriatal 6-OHDA reduced amphetamine-induced rotational behavior and loss of substantia nigra dopamine (DA) neurons. BBJ consumption alone had no ameliorative effects, but when combined with exercise, behavioral deficits and nigrostriatal DA neurodegeneration were reduced to a greater extent than exercise alone. The neuroprotection observed with exercise alone was associated with an increase in striatal glial cell-lined derived neurotrophic factor (GDNF), whereas combining exercise and BBJ was associated with an increase in nigral GDNF. These results suggest that polyphenols may potentiate the protective effects of exercise and that differential regulation of GDNF expression underlies protection observed with exercise alone versus combined treatment with consumption of BBJ.

2.
CNS Neurosci Ther ; 27(5): 528-539, 2021 05.
Article in English | MEDLINE | ID: mdl-33650762

ABSTRACT

Microglia are important phagocytes of the central nervous system (CNS). They play an important role in protecting the CNS by clearing necrotic tissue and apoptotic cells in many CNS diseases. However, recent studies have found that microglia can phagocytose parts of neurons excessively, such as the neuronal cell body, synapse, or myelin sheaths, before or after the onset of CNS diseases, leading to aggravated injury and impaired tissue repair. Meanwhile, reduced phagocytosis of synapses and myelin results in abnormal circuit connections and inhibition of remyelination, respectively. Previous studies focused primarily on the positive effects of microglia phagocytosis, whereas only a few studies have focused on the negative effects. In this review, we use the term "pathological microglial phagocytosis" to refer to excessive or reduced phagocytosis by microglia that leads to structural or functional abnormalities in target cells and brain tissue. The classification of pathological microglial phagocytosis, the composition, and activation of related signaling pathways, as well as the process of pathological phagocytosis in various kinds of CNS diseases, are described in this review. We hypothesize that pathological microglial phagocytosis leads to aggravation of tissue damage and negative functional outcome. For example, excessive microglial phagocytosis of synapses can be observed in Alzheimer's disease and schizophrenia, leading to significant synapse loss and memory impairment. In Parkinson's disease, ischemic stroke, and traumatic brain injury, excessive microglial phagocytosis of neuronal cell bodies causes impaired gray matter recovery and sensory dysfunction. We therefore believe that more studies should focus on the mechanism of pathological microglial phagocytosis and activation to uncover potential targets of therapeutic intervention.


Subject(s)
Central Nervous System Diseases/pathology , Microglia/pathology , Phagocytosis , Animals , Humans , Myelin Sheath , Neurons/pathology , Synapses/pathology
3.
Appl Energy ; 304: 117711, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-36568493

ABSTRACT

Shelter-in-place orders and business closures related to COVID-19 changed the hourly profile of electricity demand and created an unprecedented source of uncertainty for the grid. The potential for continued shifts in electricity profiles has implications for electricity sector investment and operating decisions that maintain reserve margins and provide grid reliability. This study reveals that understanding this uncertainty requires an understanding of the underlying drivers at the customer-class scale. This paper utilizes three datasets to compare the impacts of COVID-19 on electricity consumption across a range of spatiotemporal and customer scales. At the utility/customer-class scale, COVID-19-induced shutdowns in the spring of 2020 shifted weekday residential load profiles to resemble weekend profiles from previous years. Total commercial loads declined, but the commercial diurnal load profile was unchanged. With only total loads available at the balancing authority scale, the apparent impact of COVID-19 was smaller during the summer due in part to phased re-opening and spatial variability in re-opening, but there were still clear variations once total loads were broken down zonally. Monthly data at the state scale showed an increase in state-level residential electricity sales, a decrease in commercial sales, and a small net decrease in total sales in most states from April-August 2020. Analyses that focus on total load or a single scale may miss important changes that become apparent when the load is broken down regionally or by customer class.

4.
CNS Neurosci Ther ; 26(12): 1219-1229, 2020 12.
Article in English | MEDLINE | ID: mdl-33210839

ABSTRACT

Demyelination occurs in response to brain injury and is observed in many neurodegenerative diseases. Myelin is synthesized from oligodendrocytes in the central nervous system, and oligodendrocyte death-induced demyelination is one of the mechanisms involved in white matter damage after stroke and neurodegeneration. Oligodendrocyte precursor cells (OPCs) exist in the brain of normal adults, and their differentiation into mature oligodendrocytes play a central role in remyelination. Although the differentiation and maturity of OPCs drive endogenous efforts for remyelination, the failure of axons to remyelinate is still the biggest obstacle to brain repair after injury or diseases. In recent years, studies have made attempts to promote remyelination after brain injury and disease, but its cellular or molecular mechanism is not yet fully understood. In this review, we discuss recent studies examining the demyelination process and potential therapeutic strategies for remyelination in aging and stroke. Based on our current understanding of the cellular and molecular mechanisms underlying remyelination, we hypothesize that myelin and oligodendrocytes are viable therapeutic targets to mitigate brain injury and to treat demyelinating-related neurodegeneration diseases.


Subject(s)
Aging/metabolism , Brain/metabolism , Demyelinating Diseases/metabolism , Myelin Sheath/metabolism , Stroke/metabolism , Aging/pathology , Animals , Brain/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Demyelinating Diseases/pathology , Demyelinating Diseases/therapy , Humans , Myelin Sheath/pathology , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Stroke/pathology , Stroke/therapy
5.
CNS Neurosci Ther ; 25(12): 1329-1342, 2019 12.
Article in English | MEDLINE | ID: mdl-31697040

ABSTRACT

AIMS: Blood-borne monocytes/macrophages infiltrate the brain in massive numbers after ischemic stroke, but their impact on poststroke brain injury and recovery remains elusive. This study examined the transcriptomic changes in monocytes/macrophages after ischemic stroke and the functional implications of these changes, particularly with regards to the contribution of these cells to the phagocytic clearance of dead/dying cells (efferocytosis) in the poststroke brain. METHODS: We performed whole-genome RNA sequencing on the monocyte/macrophage population sorted from mouse brain and peripheral blood 5 days after permanent focal cerebral ischemia. In addition, the spatial and temporal profiles of macrophage efferocytosis were examined in vivo by immunohistochemistry 3-7 days after brain ischemia. RESULTS: Robust transcriptomic changes occurred in monocytes/macrophages upon infiltrating the poststroke brain. Functional enrichment analysis revealed a transcriptome of brain macrophages that strongly favored efferocytic activity. A large number of efferocytosis-related genes were upregulated in brain macrophages, the products of which are essential components involved in various steps of efferocytosis, such as chemotaxis, recognition of dead cells, engulfment, and processing of phagosomes. The efferocytic activity of brain macrophages were verified by immunohistochemistry, wherein Iba1-labeled microglia/macrophages effectively cleared apoptotic neurons in the infarct during the subacute stage after brain ischemia. We also identified PPARγ and STAT6 as potential upstream regulators that shaped this proefferocytic and inflammation-resolving transcriptome of macrophages in the poststroke brain. CONCLUSION: Macrophages play a crucial role in the phagocytic clearance of dead neurons after ischemic stroke and promote the resolution of inflammation in the brain. Molecular therapies that enhance macrophage efferocytic capability may be promising treatments for ischemic stroke by facilitating inflammation resolution, brain repair, and recovery of neurological functions.


Subject(s)
Brain Ischemia/pathology , Brain/pathology , Inflammation/pathology , Macrophages/pathology , Stroke/pathology , Animals , Apoptosis , Immunohistochemistry , Inflammation/etiology , Male , Mice , Mice, Inbred C57BL , Monocytes/pathology , Neuroimaging , PPAR gamma/genetics , Phagocytosis , STAT6 Transcription Factor/genetics , Sequence Analysis, RNA , Transcriptome , Whole Genome Sequencing
6.
Neurobiol Dis ; 126: 62-75, 2019 06.
Article in English | MEDLINE | ID: mdl-30218758

ABSTRACT

Post-stroke treatment with omega-3 polyunsaturated fatty acids (n-3 PUFAs) may be a promising therapy in young animals but this has not been tested in aged subjects, a population at most risk of ischemic stroke. Herein we examined the therapeutic efficacy of n-3 PUFAs after distal middle cerebral artery occlusion (dMCAO) in young (10-12 weeks old) and aged (18 months old) mice. Post-ischemic mice were randomly assigned to 4 groups that received: 1) regular food with low content of n-3 PUFAs, 2) intraperitoneal docosahexaenoic acid (DHA, a major component of n-3 PUFAs) injections, 3) Fish oil (FO, containing high concentration of n-3 PUFAs) dietary supplement, or 4) combined treatment with DHA and FO dietary supplement. Long-term neurorestoration induced by n-3 PUFA post-stroke administration and its underlying mechanism(s) were analyzed up to 35 days after dMCAO. Aged mice showed more severe neurological deficits than young mice after dMCAO with histological lesions extended to the striatum. Notably, post-stroke treatment with combined DHA injections and FO dietary supplementation was more effective in reducing brain injury and improving sensorimotor function in aged mice than either treatment alone, albeit to a lesser extent than in the young mice. Unlike the improvement in spatial cognitive function observed in young mice, the combined treatment regimen failed to improve cognitive function in aged mice. The reduction in stroke-induced neurological deficits with n-3 PUFA post-treatment was associated with enhanced angiogenesis, oligodendrogenesis, neuron survival and white matter restoration. Together, these results indicate that the neurological benefits of n-3 PUFA administration after stroke extend to older animals and are associated with improved neuronal survival and brain remodeling, therefore suggesting that post-stroke administration of n-3 PUFAs is a viable clinically relevant treatment option against stroke.


Subject(s)
Aging , Brain/drug effects , Fatty Acids, Omega-3/pharmacology , Neovascularization, Physiologic/drug effects , Stroke/pathology , Animals , Male , Mice , Mice, Inbred C57BL , Random Allocation
7.
J Biol Chem ; 291(34): 17510-22, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27365397

ABSTRACT

We previously reported that the vesicular monoamine transporter 2 (VMAT2) is physically and functionally coupled with Hsc70 as well as with the dopamine synthesis enzymes tyrosine hydroxylase (TH) and aromatic amino acid decarboxylase, providing a novel mechanism for dopamine homeostasis regulation. Here we expand those findings to demonstrate that Hsc70 physically and functionally interacts with TH to regulate the enzyme activity and synaptic vesicle targeting. Co-immunoprecipitation assays performed in brain tissue and heterologous cells demonstrated that Hsc70 interacts with TH and aromatic amino acid decarboxylase. Furthermore, in vitro binding assays showed that TH directly binds the substrate binding and carboxyl-terminal domains of Hsc70. Immunocytochemical studies indicated that Hsc70 and TH co-localize in midbrain dopaminergic neurons. The functional significance of the Hsc70-TH interaction was then investigated using TH activity assays. In both dopaminergic MN9D cells and mouse brain synaptic vesicles, purified Hsc70 facilitated an increase in TH activity. Neither the closely related protein Hsp70 nor the unrelated Hsp60 altered TH activity, confirming the specificity of the Hsc70 effect. Overexpression of Hsc70 in dopaminergic MN9D cells consistently resulted in increased TH activity whereas knockdown of Hsc70 by short hairpin RNA resulted in decreased TH activity and dopamine levels. Finally, in cells with reduced levels of Hsc70, the amount of TH associated with synaptic vesicles was decreased. This effect was rescued by addition of purified Hsc70. Together, these data demonstrate a novel interaction between Hsc70 and TH that regulates the activity and localization of the enzyme to synaptic vesicles, suggesting an important role for Hsc70 in dopamine homeostasis.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/metabolism , HSC70 Heat-Shock Proteins/metabolism , Synaptic Vesicles/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Cell Line , Chaperonin 60/genetics , Chaperonin 60/metabolism , Dopamine/genetics , Dopaminergic Neurons/cytology , HSC70 Heat-Shock Proteins/genetics , Homeostasis/physiology , Male , Mice , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Protein Binding/physiology , Protein Domains , Rats , Rats, Sprague-Dawley , Synaptic Vesicles/genetics , Tyrosine 3-Monooxygenase/genetics
8.
Exp Brain Res ; 234(7): 1863-1873, 2016 07.
Article in English | MEDLINE | ID: mdl-26894890

ABSTRACT

Insulin-like growth factor-1 (IGF-1) is an endogenous peptide transported across the blood brain barrier that is protective in several brain injury models, including an acute animal model of Parkinson's disease (PD). Motor deficits in PD are due largely to the progressive loss of nigrostriatal dopaminergic neurons. Thus, we examined the neuroprotective potential of IGF-1 in a progressive model of dopamine deficiency in which 6-hydroxydopamine (6-OHDA) is infused into the striatum. Rats received intrastriatal IGF-1 (5 or 50 µg) 6 h prior to infusion of 4 µg 6-OHDA into the same site and were euthanized 1 or 4 weeks later. Both concentrations of IGF-1 protected tyrosine hydroxylase (TH) immunoreactive terminals in striatum at 4 weeks but not at 1 week, indicating that IGF-induced restoration of the dopaminergic phenotype occurred over several weeks. TH-immunoreactive cell loss was only attenuated with 50 µg IGF-1. We then examined the effect of striatal IGF-1 on the Ras/ERK1/2 and PI3K/Akt pathways to ascertain whether their activation correlated with IGF-1-induced protection. Striatal and nigral levels of phospho-ERK1/2 were maximal 6 h after IGF-1 infusion and, with the exception of an increase in nigral pERK2 at 48 h, returned to basal levels by 7 days. Phospho-Akt (Ser473) was elevated 6-24 h post-IGF-1 infusion in both striatum and substantia nigra concomitant with inhibition of pro-death GSK-3ß, a downstream target of Akt. These results suggest that IGF-1 can protect the nigrostriatal pathway in a progressive PD model and that this protection is preceded by activation of key pro-survival signaling cascades.


Subject(s)
Dopaminergic Neurons/metabolism , Insulin-Like Growth Factor I/pharmacology , Neostriatum/metabolism , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/metabolism , Substantia Nigra/metabolism , Adrenergic Agents/administration & dosage , Adrenergic Agents/pharmacology , Animals , Disease Models, Animal , Glycogen Synthase Kinase 3 beta/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Insulin-Like Growth Factor I/administration & dosage , MAP Kinase Signaling System/drug effects , Male , Neostriatum/drug effects , Neuroprotective Agents/administration & dosage , Oxidative Stress , Oxidopamine/administration & dosage , Oxidopamine/pharmacology , Parkinsonian Disorders/drug therapy , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Tyrosine 3-Monooxygenase/drug effects , Tyrosine 3-Monooxygenase/metabolism
9.
J Learn Disabil ; 48(5): 511-22, 2015.
Article in English | MEDLINE | ID: mdl-24300589

ABSTRACT

Children with specific learning disabilities (SLD) have deficits in the basic psychological processes that interfere with learning and academic achievement, and for some SLD subtypes, these deficits can also lead to emotional and/or behavior problems. This study examined psychosocial functioning in 123 students, aged 6 to 11, who underwent comprehensive evaluations for learning and/or behavior problems in two Pacific Northwest school districts. Using concordance-discordance model (C-DM) processing strengths and weaknesses SLD identification criteria, results revealed working memory SLD (n = 20), processing speed SLD (n = 30), executive SLD (n = 32), and no disability groups (n = 41). Of the SLD subtypes, repeated measures MANOVA results revealed the processing speed SLD subtype exhibited the greatest psychosocial and adaptive impairment according to teacher behavior ratings. Findings suggest processing speed deficits may be behind the cognitive and psychosocial disturbances found in what has been termed "nonverbal" SLD. Limitations, implications, and future research needs are addressed.


Subject(s)
Adaptation, Psychological/physiology , Child Behavior/psychology , Learning Disabilities/physiopathology , Problem Behavior/psychology , Social Adjustment , Child , Female , Humans , Learning Disabilities/classification , Male
10.
Brain Res ; 1370: 80-8, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21062624

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) protects dopamine (DA) neurons from 6-hydroxydopamine (6-OHDA) toxicity. We have now explored this protection over 8 weeks following toxin administration. Infusion of Fluoro-Gold (FG) into the striatum was followed 1 week later by GDNF (9µg) or its vehicle. Six hours later, animals received 6-OHDA (4 µg) into the same site. 6-OHDA caused a loss of cells in the substantia nigra that expressed both FG and tyrosine hydroxylase (TH) and striatal terminals expressing TH, the high affinity dopamine transporter (DAT), and the vesicular monoamine transporter 2 (VMAT2) as assessed 2-8 weeks later. Loss of FG(+) cells, and striatal DA was completely blocked by GDNF by 2 weeks. In contrast, GDNF only slightly attenuated the loss of TH, DAT, or VMAT2 in the striatum at 2 weeks, but had restored these markers by 4-8 weeks. Thus, GDNF prevents DA cell death and loss of striatal DA content, but several weeks are required to fully restore the dopaminergic phenotype. These results provide insight into the mechanism of GDNF protection of DA neurons, and may help avoid incorrect interpretations of temporary phenotypic changes.


Subject(s)
Corpus Striatum/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Substantia Nigra/drug effects , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Male , Neural Pathways/drug effects , Neural Pathways/metabolism , Neural Pathways/pathology , Neuroprotective Agents/therapeutic use , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/physiopathology , Rats , Rats, Sprague-Dawley , Substantia Nigra/metabolism , Substantia Nigra/pathology
11.
J Neurosci Methods ; 191(2): 234-8, 2010 Aug 30.
Article in English | MEDLINE | ID: mdl-20600314

ABSTRACT

Experiments often involve multiple analyses, such as assays of neurotransmitters and proteins, and this can require different initial sample preparations. Typically, this is accomplished by using different animals or different tissue samples from the same animal. Either approach renders comparisons between assays more variable and greatly increases the effort and/or cost. Using tissue collected from rat striatum and molecules of special relevance to studies of Parkinson's disease, we show that tissue sonication in water prior to aliquoting into the appropriate concentrated solutions (e.g. HClO(4) and lysis buffers) permits several types of measurements to be made from the same initial samples. Dopamine and its metabolite homovanillic acid, serotonin and its metabolite 5-hydroxyindoleacetic acid, tyrosine hydroxylase and its phosphorylation at Ser19 and Ser31, and the dopamine transporter were unaffected. However, phospho-Akt levels fell slightly and phospho-ERK1/2 tended to drop. We also present a simple technique to preserve phosphorylation state of proteins such as ERK1/2 by perfusing animals through the heart with a phosphatase inhibitor, NaF. Dopamine metabolite dihydroxyphenyl acetic acid (DOPAC) levels were raised with both techniques, however. The general principles reported here are likely to apply to other brain regions, facilitate multiple comparisons of variables, increase efficiency, and decrease costs.


Subject(s)
Brain Chemistry/physiology , Brain/metabolism , Neurochemistry/methods , Sonication/methods , Animals , Biological Assay/methods , Corpus Striatum/chemistry , Corpus Striatum/metabolism , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley
12.
Alcohol Clin Exp Res ; 33(12): 2113-23, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19740131

ABSTRACT

BACKGROUND: The purpose of the present investigation was to more clearly define blood-alcohol parameters associated with alcohol dependence produced by alcohol vapor inhalation and alcohol-containing liquid diet. METHODS: Alcohol levels in blood and brain were compared during and after 4 hours of acute alcohol vapor exposure; also, brain-alcohol levels were assessed in alcohol-exposed (14-day alcohol vapor) and alcohol-naïve rats during and after 4 hours of acute alcohol vapor exposure. A separate group of rats were implanted with i.v. catheters, made dependent on alcohol via vapor inhalation, and tested for operant alcohol responding; blood-alcohol levels (BALs) were measured throughout operant alcohol drinking sessions during alcohol withdrawal. A final group of rats consumed an alcohol-liquid diet until they were dependent, and those rats were then tested for operant behavior at various withdrawal time points; BALs were measured at different withdrawal time points and after operant sessions. RESULTS: Blood- and brain-alcohol levels responded similarly to vapor, but brain-alcohol levels peaked at a higher point and more slowly returned to zero in alcohol-naïve rats relative to alcohol-exposed rats. Alcohol vapor exposure also produced an upward shift in subsequent operant alcohol responding and resultant BALs. Rats consumed large quantities of alcohol-liquid diet, most of it during the dark cycle, sufficient to produce high blood-alcohol levels and elevated operant alcohol responding when tested during withdrawal from liquid diet. CONCLUSIONS: These results emphasize that the key determinants of excessive alcohol drinking behavior are the BAL range and pattern of chronic high-dose alcohol exposure.


Subject(s)
Alcoholism/metabolism , Alcoholism/psychology , Central Nervous System Depressants/metabolism , Central Nervous System Depressants/pharmacology , Conditioning, Operant/drug effects , Ethanol/metabolism , Ethanol/pharmacology , Administration, Inhalation , Animals , Brain/metabolism , Central Nervous System Depressants/blood , Diet , Ethanol/blood , Injections, Intravenous , Male , Microdialysis , Rats , Rats, Wistar , Self Administration
13.
J Biol Chem ; 284(21): 14011-9, 2009 May 22.
Article in English | MEDLINE | ID: mdl-19289463

ABSTRACT

The 14-3-3 proteins stimulate the activation of tyrosine hydroxylase (TH), the rate-limiting catecholamine biosynthetic enzyme. To explore if particular endogenous 14-3-3 isoforms specifically affected TH activity and dopamine synthesis, we utilized rodent nigrostriatal tissues and midbrain-derived MN9D dopaminergic cells. Extracts from ventral midbrain and MN9D cells contained similar pools of 14-3-3 mRNAs, with 14-3-3zeta being relatively abundant in both. Protein levels of 14-3-3zeta were also abundant. [(32)P]Orthophosphate labeling of MN9D cells, followed by co-immunoprecipitation with pan-TH and pan-14-3-3 antibodies brought down similar amounts of phosphorylated TH in each, confirming that 14-3-3-bound phosphorylated TH in our cells. Co-immunoprecipitation of striatal tissues with a pan-TH antibody precipitated 14-3-3zeta but not another potential TH regulatory isoform, 14-3-3eta. In whole cell extracts from MN9D cells after 14-3-3 small interfering RNA treatments, we found that 14-3-3zeta knockdown significantly reduced TH activity and dopamine synthesis whereas knockdown of 14-3-3eta had no effect. 14-3-3zeta was found co-localized on mitochondria with TH, and its knockdown by small interfering RNA reduced TH phosphorylation and TH activity in the mitochondrial pool. Together the data support a role for 14-3-3zeta as an endogenous activator of TH in midbrain dopaminergic neurons and furthermore, identify mitochondria as a potential novel site for dopamine synthesis, with implications for Parkinson disease.


Subject(s)
14-3-3 Proteins/metabolism , Dopamine/metabolism , Mitochondria/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Cell Line , Dihydroxyphenylalanine/metabolism , Gene Expression Regulation , Immunoprecipitation , Mesencephalon/cytology , Mice , Mitochondria/enzymology , Mitochondria/ultrastructure , Neurons/metabolism , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats
14.
Parkinsonism Relat Disord ; 15 Suppl 3: S42-5, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20083005

ABSTRACT

We are testing the hypothesis that exercise is neuroprotective in animal models of the dopamine (DA) deficiency in Parkinson's disease. Our studies include mice or rats provided access to a running wheel and subsequently treated with MPTP (mice) or 6-hydroxydopamine (rats) and monkeys provided access to a treadmill and subsequently treated with MPTP. Typically, the exercise occurs for 3 months prior to the toxin treatment and often for 1-2 months thereafter. Our findings indicate that exercise reduces the behavioral impairments elicited by the dopaminergic neurotoxins as well as the loss of DA neurons as assessed by PET imaging and biochemical or histochemical assessment of tissue samples. Our studies are focused on one of several possible explanations for the beneficial effects of exercise: an exercise-induced increase in the expression of neurotrophic factors, particularly GDNF. Our observations indicate that GDNF can reduce the vulnerability of DA neurons, in part due to the activation of key intracellular cascades. We also find that mild cellular stress itself can provide protection against more intensive stress, a form of preconditioning. We conclude that dopamine neurons have the capacity to respond to intracellular and extracellular signals by triggering endogenous neuroprotective mechanisms. This raises the possibility that some individuals with Parkinson's disease suffer from a reduction of these neuroprotective mechanisms, and that treatments that boost these mechanisms - including exercise - may provide therapeutic benefit.


Subject(s)
Dopamine/deficiency , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/rehabilitation , Physical Conditioning, Animal/methods , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Mice , Neurons/metabolism , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Rats
15.
J Neurosci Res ; 86(9): 2039-49, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18438911

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) has been shown to be neuroprotective in animal models of the dopamine deficiency in Parkinson's disease. To examine the role of the extracellular signal-regulated kinases 1 and 2 (ERK1/2) in this process, we infused a single dose of GDNF into the striatum of mice and analyzed the effect on ERK1/2 by immunohistochemistry and Western blot analysis. GDNF caused an increase in the phosphorylation of ERK1/2 both in the striatum and in tyrosine hydroxylase-positive neurons in the substantia nigra. In the striatum, the increase in ERK1/2 phosphorylation was evident by 3 hr and persisted for at least 7 days, whereas, in the substantia nigra, an increase in phosphorylated ERK1/2 was first evident at 24 hr and persisted for at least 7 days. The increase in phosphorylated ERK1/2 was maximal at 0.45 microg GDNF at the time points examined. GDNF also protected dopamine terminals against the loss of tyrosine hydroxylase immunoreactivity normally associated with the intrastriatal administration of 6-hydroxydopamine (0.5 microg/0.5 microl). However, this was observed only at a much higher dose of GDNF, 4.5 microg. Thus, our results suggest that the ability of GDNF to protect dopamine neurons cannot be explained solely in terms of its influence on ERK1/2 and that the role of other signaling pathways should be explored.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neuroglia/enzymology , Parkinsonian Disorders/physiopathology , Animals , Disease Models, Animal , Enzyme Activation , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 3/drug effects , Oxidopamine/antagonists & inhibitors , Oxidopamine/toxicity , Parkinsonian Disorders/enzymology
16.
J Neurochem ; 102(6): 2118-2131, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17555551

ABSTRACT

Activated microglia are an important feature of many neurological diseases and can be imaged in vivo using 1-(2-chlorophenyl)-N-methyl-N-(1-methylpropyl)-3-isoquinolinecarboxamide (PK11195), a ligand that binds the peripheral benzodiazepine receptor (PBR). N-(2,5-dimethoxybenzyl)-N-(5-fluoro-2-phenoxyphenyl) acetamide (DAA1106) is a new PBR-specific ligand that has been reported to bind to PBR with higher affinity compared with PK11195. We hypothesized that this high-affinity binding of DAA1106 to PBR will enable better delineation of microglia in vivo using positron emission tomography. [(3)H]DAA1106 showed higher binding affinity compared with [(3)H](R)-PK11195 in brain tissue derived from normal rats and the rats injected intrastriatally with 6-hydroxydopamine or lipopolysaccharide at the site of the lesion. Immunohistochemistry combined with autoradiography in brain tissues as well as correlation analyses showed that increased [(3)H]DAA1106 binding corresponded mainly to activated microglia. Finally, ex vivo autoradiography and positron emission tomography imaging in vivo showed greater retention of [(11)C]DAA1106 compared with [(11)C](R)-PK11195 in animals injected with either lipopolysaccaride or 6-hydroxydopamine at the site of lesion. These results indicate that DAA1106 binds with higher affinity to microglia in rat models of neuroinflammation when compared with PK11195, suggesting that [(11)C]DAA1106 may represent a significant improvement over [(11)C](R)-PK11195 for in vivo imaging of activated microglia in human neuroinflammatory disorders.


Subject(s)
Acetamides , Binding, Competitive/physiology , Encephalitis/diagnostic imaging , Isoquinolines , Microglia/drug effects , Phenyl Ethers , Receptors, GABA-A/drug effects , Acetamides/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Brain/physiopathology , Disease Models, Animal , Encephalitis/metabolism , Encephalitis/physiopathology , Gliosis/diagnostic imaging , Gliosis/metabolism , Gliosis/physiopathology , Isoquinolines/metabolism , Ligands , Lipopolysaccharides , Male , Microglia/metabolism , Oxidopamine , Phenyl Ethers/metabolism , Positron-Emission Tomography/methods , Radioligand Assay , Rats , Receptors, GABA-A/metabolism , Tritium
17.
Biochem Pharmacol ; 70(9): 1371-81, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16144694

ABSTRACT

TEMPOL (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl) is a stable nitroxyl antioxidant. Previous studies have suggested that TEMPOL is protective in acute disorders thought to involve reactive oxygen species (ROS), such as ischemic stroke and cardiac reperfusion injury. Oxidized TEMPOL can be recycled to its redox-active reducing form by co-administration with polynitroxylated albumin, making it a candidate as a pharmacological "reservoir" for reducing potential of use in chronic disorders involving ROS. The present studies examine the efficacy of TEMPOL in cell culture and animal models of the central and peripheral dysfunction associated with Parkinson's disease, a disorder in the pathogenesis of which ROS generated from dopamine have been implicated. Antioxidants have been proposed as both preventive and symptomatic therapy for Parkinson's disease. TEMPOL protects MN9D dopaminergic mesencephalic cells in culture from 6-hydroxydopamine (6-OHDA)-induced apoptosis. Translocation of the p65 component of NF-kappaB to the nucleus accompanies protection by TEMPOL. In vivo, intraperitoneal TEMPOL protects mice from intrastriatal 6-OHDA-induced cell and dopamine metabolite loss in the striatum. TEMPOL also protects mice against the 6-OHDA-induced rotational behavior elicited by intrastriatal administration of d-amphetamine. In addition, TEMPOL protects mice from the ptosis, activity level decrement, and mortality induced by intraperitoneal administration of 6-OHDA, a model of autonomic dysfunction in Parkinson's disease. Adjunctive use of polynitroxylated albumin enhances the in vitro and in vivo effects of TEMPOL.


Subject(s)
Cyclic N-Oxides/pharmacology , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Active Transport, Cell Nucleus/drug effects , Animals , Autonomic Nervous System/drug effects , Autonomic Nervous System/physiology , Cell Line , Cell Survival/drug effects , Corpus Striatum/drug effects , Corpus Striatum/pathology , Dopamine/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Oxidopamine/toxicity , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology , Peptide Nucleic Acids/pharmacology , Reactive Oxygen Species , Spin Labels , Substantia Nigra/drug effects , Substantia Nigra/pathology
18.
Exp Neurol ; 184(1): 31-9, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14637076

ABSTRACT

Evidence suggests that following injury the brain has the capacity for self-repair and that this can be promoted through a variety of experiences including motor activity. In their article, Döbrössy and Dunnett have provided further evidence that this is the case in an animal model in which an excitotoxin is applied to the neostriatum. Under standard conditions, such a toxin would cause considerable damage to the GABAergic cells of this region and produce behavioral deficits. This model has been used to explore certain aspects of Huntington's disease, which also involves the loss of these neurons. However, Döbrössy and Dunnett show that the damage can be reduced by prior motor training. We have been exploring the neuroprotective effects of motor exercise in a different model, one involving 6-hydroxydopamine, which normally destroys dopamine neurons. Our results indicate that forced exercise can reduce the vulnerability of dopamine neurons to 6-hydroxydopamine. The results further suggest that this protection is due in part to an increase in the availability of the trophic factor GDNF, which can in turn stimulate certain signaling cascades, including one that activates ERK. Our results, together with those of Döbrössy and Dunnett and others, raise the possibility that exercise will protect against a variety of neurodegenerative conditions.


Subject(s)
Parkinson Disease, Secondary/prevention & control , Physical Conditioning, Animal/physiology , Animals , Glial Cell Line-Derived Neurotrophic Factor , Humans , Nerve Growth Factors/physiology , Oxidopamine/toxicity , Parkinson Disease, Secondary/chemically induced , Physical Exertion/physiology , Sympatholytics/toxicity , Synaptic Transmission/drug effects
19.
Brain Res Mol Brain Res ; 117(2): 129-38, 2003 Oct 07.
Article in English | MEDLINE | ID: mdl-14559146

ABSTRACT

Exogenous GDNF as well as vectors containing the gene for this trophic factor has been shown to be neuroprotective in animal models of Parkinson's disease. We therefore investigated whether changes in striatal GDNF protein and nigral mRNA levels of its co-receptors GFRalpha1 and RET occur in response to lesions of dopamine (DA) neurons and examined the temporal profile of these changes as they relate to the loss of dopaminergic markers. Rats were lesioned with 6-hydroxydopamine and sacrificed 3 h to 60 days post-infusion. DA tissue levels in the striatum and tyrosine hydroxylase immunoreactivity in the substantia nigra (SN) and ventral tegmental area (VTA) were used to determine the size of the lesions. GDNF protein was measured in the striatum using radioimmunocytochemistry. In situ hybridization was used to determine alterations in the mRNAs of RET and GFRalpha1 in the SN and VTA. We observed no persistent changes in GDNF protein in the striatum in response to 6-hydroxydopamine over the 60-day observation period, suggesting that compensatory changes in this trophic factor do not occur in response to injury. Dramatic decreases in RET and GFRalpha1 were observed in both SN and VTA that were generally correlated with the loss of TH protein and striatal DA content, strongly suggesting that these receptors are located on DA neurons and that the protective effect of GDNF reflects a direct action of the trophic factor on these neurons.


Subject(s)
Corpus Striatum/drug effects , Drosophila Proteins/metabolism , Nerve Growth Factors/metabolism , Oxidopamine/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Substantia Nigra/drug effects , Animals , Autoradiography , Chromatography, High Pressure Liquid , Corpus Striatum/metabolism , Drosophila Proteins/genetics , Functional Laterality , Glial Cell Line-Derived Neurotrophic Factor , Imaging, Three-Dimensional , Immunohistochemistry , In Situ Hybridization , Male , Medial Forebrain Bundle/drug effects , Nerve Growth Factors/genetics , Proto-Oncogene Proteins c-ret , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Substantia Nigra/metabolism , Sympatholytics/pharmacology , Time Factors , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
20.
J Neurochem ; 85(2): 299-305, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12675906

ABSTRACT

Unilateral administration of 6-hydroxydopamine (6-OHDA) into the medial forebrain bundle (MFB) causes a loss of dopamine (DA) in the ipsilateral striatum and contralateral motor deficits. However, if a cast is placed on the ipsilateral limb during the first 7 days following 6-OHDA infusion, forcing the animal to use its contralateral limb, both the behavioral and neurochemical deficits are reduced. Here, we examine the effect of forced reliance on a forelimb during the 7 days prior to ipsilateral infusion of 6-OHDA on the deficits characteristic of this lesion model. Casted animals displayed no behavioral asymmetries as measured 14-28 days postlesion and a marked attenuation in the loss of striatal DA and its metabolites at 30 days. In addition, animals receiving a unilateral cast alone had an increase in glial cell-line derived neurotrophic factor (GDNF) protein in the striatum corresponding to the overused limb. GDNF increased within 1 day after the onset of casting, peaked at 3 days, and returned to baseline within 7 days. These results suggest that preinjury forced limb-use can prevent the behavioral and neurochemical deficits to the subsequent administration of 6-OHDA and that this may be due in part to neuroprotective effects of GDNF.


Subject(s)
Forelimb , Medial Forebrain Bundle/drug effects , Nerve Growth Factors/metabolism , Oxidopamine/pharmacology , Parkinson Disease, Secondary/physiopathology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Apomorphine/pharmacology , Behavior, Animal/drug effects , Casts, Surgical , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Forelimb/physiopathology , Functional Laterality , Glial Cell Line-Derived Neurotrophic Factor , Immobilization/physiology , Male , Motor Activity/drug effects , Oxidopamine/administration & dosage , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/prevention & control , Rats , Rats, Long-Evans , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...