Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 48
1.
PLoS One ; 16(2): e0245663, 2021.
Article En | MEDLINE | ID: mdl-33534843

Parkinson's disease is associated with the loss of dopamine (DA) neurons in ventral mesencephalon. We have previously reported that no single neurotrophic factor we tested protected DA neurons from the dopaminergic toxin 1-methyl-4-phenylpyridinium (MPP+) in dissociated cultures isolated from the P0 rat substantia nigra, but that a combination of five neurotrophic factors was protective. We now report that cerebral DA neurotrophic factor (CDNF) and a variant of neurturin (NRTN), N4, were also not protective when provided alone but were protective when added together. In cultures isolated from the substantia nigra, MPP+ (10 µM) decreased tyrosine hydroxylase-positive cells to 41.7 ± 5.4% of vehicle control. Although treatment of cultures with 100 ng/ml of either CDNF or N4 individually before and after toxin exposure did not significantly increase survival in MPP+-treated cultures, when the two trophic factors were added together at 100 ng/ml each, survival of cells was increased 28.2 ± 6.1% above the effect of MPP+ alone. In cultures isolated from the ventral tegmental area, another DA rich area, a higher dose of MPP+ (1 mM) was required to produce an EC50 in TH-positive cells but, as in the substantia nigra, only the combination of CDNF and N4 (100 ng/ml each) was successful at increasing the survival of these cells compared to MPP+ alone (by 22.5 ± 3.5%). These data support previous findings that CDNF and N4 may be of therapeutic value for treatment of PD, but suggest that they may need to be administered together.


Dopaminergic Neurons/metabolism , Mesencephalon/metabolism , Nerve Growth Factors/pharmacology , Neuroprotective Agents/pharmacology , Neurturin/pharmacology , 1-Methyl-4-phenylpyridinium , Animals , CHO Cells , Cell Survival/drug effects , Cells, Cultured , Cricetulus , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/drug effects , Humans , Nomifensine/pharmacology , Rats, Sprague-Dawley , Substantia Nigra/cytology , Tritium/metabolism , Tyrosine 3-Monooxygenase/metabolism , Ventral Tegmental Area/cytology
3.
J Biol Chem ; 291(34): 17510-22, 2016 08 19.
Article En | MEDLINE | ID: mdl-27365397

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.


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
4.
Eur J Neurosci ; 44(1): 1691-9, 2016 07.
Article En | MEDLINE | ID: mdl-27098376

Parkinson's disease is a progressive neurodegenerative disorder, caused in part by the loss of dopamine (DA) neurons in the substantia nigra (SN). Neurotrophic factors have been shown to increase the basal survival of DA neurons in vitro, as well as to protect the neurons from some toxins under certain in vitro conditions and in animal models. Although these factors have often been tested individually, they have rarely been studied in combinations. We therefore examined the effect of such combinations after acute exposure to the toxin 1-methyl-4-phenylpyridinium (MPP(+) ) using dissociated postnatal rat midbrain cultures isolated from SN and ventral tegmental area (VTA). We found that significant loss of DA neurons in the SN occurred with an LC50 of between 1 and 10 µm, whereas the LC50 of DA neurons from the VTA was approximately 1000-fold higher. We did not observe neuroprotection against MPP(+) by individual exposure to glial cell-line derived neurotrophic factor (GDNF), brain derived neurotrophic factor (BDNF), transforming growth factor beta (TGFß), basic fibroblast growth factor (FGF-2) or growth/differentiation factor 5 (GDF5) at concentrations of 100 or 500 ng/mL. Combinations of two, three or four neurotrophic factors were also ineffective. However, when the SN cultures were exposed to a combination of all five neurotrophic factors, each at a concentration of 100 ng/mL, we observed a 30% increase in DA neuron survival in the presence of 10 and 500 µm MPP(+) . These results may be relevant to the use of neurotrophic factors as therapeutic treatments for Parkinson's disease.


1-Methyl-4-phenylpyridinium/toxicity , Dopaminergic Neurons/drug effects , Nerve Growth Factors/pharmacology , Neuroprotective Agents/pharmacology , Animals , Cell Survival , Cells, Cultured , Dopaminergic Neurons/physiology , Female , Male , Mesencephalon/cytology , Rats , Rats, Sprague-Dawley
5.
Exp Brain Res ; 234(7): 1863-1873, 2016 07.
Article En | MEDLINE | ID: mdl-26894890

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.


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
6.
Int J Dev Neurosci ; 46: 44-50, 2015 Nov.
Article En | MEDLINE | ID: mdl-26363522

Degeneration and dysfunctioning of dopaminergic neurons in the midbrain have been associated with serious neurodegenerative and neuropsychiatric disorders. Elucidating the underlying neurobiology of these neurons during early postnatal development may provide important information regarding the etiology of these disorders. Cellular signaling pathways have been shown to regulate postnatal neuronal development. Among several signaling pathways, extracellular-regulated mitogen kinases (ERK) 1, 2, and 5 have been shown to be crucial for the survival and function of dopaminergic neurons. In this study, the basal expression and activation of ERK1, 2, and 5 were studied during postnatal development in regions rich in DA cells and terminals. In the striatum (STR) and ventral mesencephalon regions of the substantia nigra (SN) and ventral tegmental area (VTA), ERK5 expression and activation were high during early postnatal days and declined with aging. Interestingly, sharp increases in phosphorylated or activated ERK1 and ERK2 were observed at postnatal day (PND) 7 in the SN and VTA. In contrast, in the STR, the levels of phosphorylated ERK1 and 2 were significantly higher at PND0 than at any other PND examined. Overall, the understanding of alterations in ERK signaling in regions rich in DA cells and DA terminals during postnatal neuronal development may provide information about their role in regulation of dopamine neuronal development which may ultimately provide insight into the underlying mechanisms of dopamine neurodegeneration.


Aging , Brain/growth & development , Brain/metabolism , Dopamine/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Developmental/physiology , Animals , Animals, Newborn , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Male , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 7 , Pregnancy , Rats , Rats, Sprague-Dawley
7.
J Neurosci Res ; 92(11): 1425-33, 2014 Nov.
Article En | MEDLINE | ID: mdl-24916473

Loss of dopamine neurons is associated with the motor deficits that occur in Parkinson's disease. Although many drugs have proven to be useful in the treatment of the symptoms of this disease, none has been shown to have a significant impact on the development of the disease. However, we believe that several neurotrophic factors have the potential to reduce its progression. Glial cell line-derived neurotrophic factor (GDNF), a member of the transforming growth factor-ß superfamily of neurotrophic factors, has been extensively studied in this regard. Less attention has been paid to growth/differentiation factor 5 (GDF5), another member of the same superfamily. This study compares GDNF and GDF5 in dissociated cultures prepared from ventral mesencephalon and in organotypic co-cultures containing substantia nigra, striatum, and neocortex. We report that both GDNF (10-500 ng/ml) and GDF5 (100-500 ng/ml) promoted the survival of dopamine neurons from the substantia nigra of postnatal rats, although GDNF was considerably more potent than GDF5. In contrast, neither factor had any significant effect on the survival of dopamine neurons from the rat ventral tegmental area. Using organotypic co-cultures, we also compared GDF5 with GDNF as chemoattractants for the innervation of the striatum and the neocortex by dopamine neurons from the substantia nigra. The addition of either GDF5 or GDNF (100-500 ng/ml) caused innervation by dopamine neurons into the cortex as well as the striatum, which did not occur in untreated cultures. Our results are consistent with similar findings suggesting that GDF5, like GDNF, deserves attention as a possible therapeutic intervention for Parkinson's disease.


Dopaminergic Neurons/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Growth Differentiation Factor 5/pharmacology , Ventral Tegmental Area/cytology , Analysis of Variance , Animals , Animals, Newborn , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cell Survival/drug effects , Cells, Cultured , Coculture Techniques , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Nerve Net/drug effects , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Substantia Nigra , Time Factors , Tyrosine 3-Monooxygenase/metabolism
8.
Neurobiol Aging ; 35(3): 669-79, 2014 Mar.
Article En | MEDLINE | ID: mdl-24411019

Extracellular signal-regulated kinases (ERKs) 1, 2, and 5 have been shown to play distinct roles in proliferation, differentiation, and neuronal viability. In this study, we examined ERK1, 2, and 5 expression and activation in the substantia nigra (SN), striatum (STR), and ventral tegmental area (VTA) during aging. An age-related decrease in phosphorylated ERK5 was observed in the SN and STR, whereas an increase in total ERK1 was observed in all 3 regions. In primary cultures of the SN and VTA, inhibition of ERK5 but not ERK1 and 2 decreased dopamine neuronal viability significantly. These data suggest that ERK5 is essential for the basal survival of SN and VTA dopaminergic neurons. This is the first study to examine ERK1, 2, and 5 expression and activation in the SN, STR, and VTA during aging, and the relative roles of ERK1, 2, and 5 in basal survival of SN and VTA dopaminergic neurons. These data raise the possibility that a decline in ERK5 signaling may play a role in age-related impairments in dopaminergic function.


Aging/pathology , Corpus Striatum/enzymology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Mitogen-Activated Protein Kinase 7/physiology , Neurons/pathology , Substantia Nigra/enzymology , Ventral Tegmental Area/enzymology , Aging/metabolism , Animals , Cell Survival/genetics , Cells, Cultured , Corpus Striatum/cytology , Corpus Striatum/pathology , Dopamine/physiology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Phosphorylation , Rats , Rats, Inbred BN , Rats, Inbred F344 , Substantia Nigra/cytology , Substantia Nigra/pathology , Ventral Tegmental Area/cytology , Ventral Tegmental Area/pathology
9.
Metab Brain Dis ; 29(2): 255-60, 2014 Jun.
Article En | MEDLINE | ID: mdl-24399497

In response to acute adversity, emotional signals shift the body into a state that permits rapid detection, identification, and appropriate response to a potential threat. The stress response involves the release of a variety of substances, including neurotransmitters, neurotrophic factors, hormones, and cytokines, that enable the body to deal with the challenges of daily life. The subsequent activation of various physiological systems can be both protective and damaging to the individual, depending on timing, intensity, and duration of the stressor. Successful recovery from stressful challenges during early life leads to strengthening of synaptic connections in health-promoting neural networks and reduced vulnerability to subsequent stressors that can be protective in later life. In contrast, chronic intense uncontrollable stress can be pathogenic and lead to disorders such as depression, anxiety, hypertension, Alzheimer's disease, Parkinson's disease, and an increased toxic response to additional stressors such as traumatic brain injury and stroke. This review briefly explores the interaction between stress experienced at different stages of development and exercise later in life.


Brain/physiology , Exercise/physiology , Nerve Net/physiology , Stress, Psychological/metabolism , Animals , Humans , Stress, Psychological/psychology
10.
Prog Neurobiol ; 114: 58-83, 2014 Mar.
Article En | MEDLINE | ID: mdl-24389580

Preconditioning is a phenomenon in which brief episodes of a sublethal insult induce robust protection against subsequent lethal injuries. Preconditioning has been observed in multiple organisms and can occur in the brain as well as other tissues. Extensive animal studies suggest that the brain can be preconditioned to resist acute injuries, such as ischemic stroke, neonatal hypoxia/ischemia, surgical brain injury, trauma, and agents that are used in models of neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. Effective preconditioning stimuli are numerous and diverse, ranging from transient ischemia, hypoxia, hyperbaric oxygen, hypothermia and hyperthermia, to exposure to neurotoxins and pharmacological agents. The phenomenon of "cross-tolerance," in which a sublethal stress protects against a different type of injury, suggests that different preconditioning stimuli may confer protection against a wide range of injuries. Research conducted over the past few decades indicates that brain preconditioning is complex, involving multiple effectors such as metabolic inhibition, activation of extra- and intracellular defense mechanisms, a shift in the neuronal excitatory/inhibitory balance, and reduction in inflammatory sequelae. An improved understanding of brain preconditioning should help us identify innovative therapeutic strategies that prevent or at least reduce neuronal damage in susceptible patients. In this review, we focus on the experimental evidence of preconditioning in the brain and systematically survey the models used to develop paradigms for neuroprotection, and then discuss the clinical potential of brain preconditioning.


Central Nervous System Diseases/therapy , Ischemic Preconditioning , Neuroprotective Agents/therapeutic use , Animals , Disease Models, Animal , Humans
11.
Parkinsonism Relat Disord ; 20 Suppl 1: S123-7, 2014 Jan.
Article En | MEDLINE | ID: mdl-24262162

There is clinical evidence that the symptoms of Parkinson's disease can be ameliorated by physical exercise, and we have been using animal models to explore the hypothesis that such exercise can also be neuroprotective. To do so we have focused on models of the dopamine deficiency associated with motor symptoms of parkinsonism, including mice treated systemically with MPTP and rats treated with 6-hydroxydopamine. Our focus on exercise derives in part from the extensive literature on the ability of exercise to increase mitochondrial respiration and antioxidant defenses, and to stimulate neuroplasticity. Beginning with constraint therapy and then employing wheel running and environmental enrichment, we have shown that increased limb use can reduce the behavioral effects of dopamine-directed neurotoxins and reduce the loss of dopamine neurons that would otherwise occur. While the mechanism of these effects is not yet known, we suspect a central role for neurotrophic factors whose expression can be stimulated by exercise and which can act on dopamine neurons to reduce their vulnerability to toxins. We believe these data, together with observations from several other laboratories, suggest that exercise, as well as neurotrophic factors, is likely to be an effective neuroprotective strategy in the treatment of Parkinson's disease.


Exercise , Parkinson Disease/rehabilitation , Animals , Disease Models, Animal , Humans , Neuroprotective Agents , Parkinson Disease/etiology
13.
J Microbiol Biol Educ ; 15(2): 83-7, 2014 Dec.
Article En | MEDLINE | ID: mdl-25574252
14.
Front Biosci (Elite Ed) ; 4(7): 2365-77, 2012 06 01.
Article En | MEDLINE | ID: mdl-22652643

The enhanced neurotoxicity of the Parkinson's disease-associated LRRK2 mutant, G2019S, than its wild-type counter-part has recently been reported. Overexpression of LRRK2 (G2019S) in cultured neural cells results in caspase-3-dependent apoptosis via a yet undefined signaling pathway. Elucidation of the mechanism underlying LRRK2 (G2019S) neurotoxicity may offer new insights into the pathogenesis of Parkinson's disease. In this study, we identified glutathione s-transferase P1 (GSTP1) as a selective target whose expression is negatively regulated at the transcriptional levels via promoter hyper-methylation by LRRK2 (G2019S). Overexpression of LRRK2 (G2019S) in the human neuronal cell line SH-SY5Y markedly suppressed the expression of GSTP1 prior to any manifestation of cell death. Moreover, shRNA-mediated knockdown of endogenous GSTP1 expression exacerbated LRRK2 (G2019S) neurotoxicity, whereas overexpression of GSTP1 protected against LRRK2 (G2019S)-induced caspase-3 activation and neuronal apoptosis. In conclusion, the results suggest a previously undefined signaling mechanism underlying the neurotoxic effect of LRRK2 (G2019S), in which LRRK2 (G2019S) triggers oxidative stress in cells and, in turn, results in caspase-dependent apoptosis at least in part by suppressing the expression of GSTP1.


Cell Death , Glutathione Transferase/metabolism , Neurons/cytology , Protein Serine-Threonine Kinases/metabolism , Base Sequence , Blotting, Western , Cell Line, Tumor , DNA Primers , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique, Indirect , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Real-Time Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
15.
Genomics ; 99(3): 144-51, 2012 Mar.
Article En | MEDLINE | ID: mdl-22227022

Obesity affects over 500 million people worldwide, and has far reaching negative health effects. Given that high body mass index (BMI) and insulin resistance are associated with alterations in many regions of brain and that physical activity can decrease obesity, we hypothesized that in Rhesus monkeys (Macaca mulatta) fed a high fat diet and who subsequently received reduced calories BMI would be associated with a unique gene expression signature in motor regions of the brain implicated in neurodegenerative disorders. In the motor cortex with increased BMI we saw the upregulation of genes involved in apoptosis, altered gene expression in metabolic pathways, and the downregulation of pERK1/2 (MAPK1), a protein involved in cellular survival. In the caudate nucleus with increased BMI we saw the upregulation of known obesity related genes (the insulin receptor (INSR) and the glucagon-like peptide-2 receptor (GLP2R)), apoptosis related genes, and altered expression of genes involved in various metabolic processes. These studies suggest that the effects of high BMI on the brain transcriptome persist regardless of two months of calorie restriction. We hypothesize that active lifestyles with low BMIs together create a brain homeostasis more conducive to brain resiliency and neuronal survival.


Body Mass Index , Caudate Nucleus/metabolism , Macaca mulatta/genetics , Mitogen-Activated Protein Kinase 1/genetics , Motor Cortex/metabolism , Neurons/metabolism , Receptor, Insulin/genetics , Animals , Apoptosis/genetics , Brain/metabolism , Caloric Restriction , Gene Expression Regulation , Glucagon-Like Peptide-2 Receptor , Metabolic Networks and Pathways/genetics , Obesity/genetics , Obesity/metabolism , Receptors, Glucagon/genetics
16.
Obesity (Silver Spring) ; 20(3): 692-8, 2012 Mar.
Article En | MEDLINE | ID: mdl-22016091

It has been established that weight gain and weight loss are heavily influenced by activity level. In this study, we hypothesized that the motor cortex exhibits a distinct physical activity-associated gene expression profile, which may underlie changes in weight associated with movement. Using DNA microarrays we profiled gene expression in the motor cortex of a group of 14 female rhesus monkeys (Macaca mulatta) with a wide range of stable physical activity levels. We found that neuronal growth factor signaling and nutrient sensing transcripts in the brain were highly correlated with physical activity. A follow-up of AKT3 expression changes (a gene at the apex of neuronal survival and nutrient sensing) revealed increased protein levels of total AKT, phosphorylated AKT, and forkhead box O3 (FOXO3), one of AKT's main downstream effectors. In addition, we successfully validated three other genes via quantitative polymerase chain reaction (qPCR) (cereblon (CRBN), origin recognition complex subunit 4-like, and pyruvate dehydrogenase 4 (PDK4)). We conclude that these genes are important in the physical activity-associated pathway in the motor cortex, and may be critical for physical activity-associated changes in body weight and neuroprotection.


Motor Activity/genetics , Motor Cortex/physiopathology , Proto-Oncogene Proteins c-akt/genetics , Animals , Female , Gene Expression Profiling , Macaca mulatta/genetics , Motor Cortex/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics
17.
Parkinsonism Relat Disord ; 18 Suppl 1: S147-50, 2012 Jan.
Article En | MEDLINE | ID: mdl-22166417

Although a good deal is known about the pathophysiology of Parkinson's disease and information is emerging about its cause, there are no pharmacological treatments shown to have a significant, sustained capacity to prevent or attenuate the condition. However, accumulating clinical evidence suggests that physical exercise can provide this much needed treatment, and studies of animal models of the dopamine deficiency associated with the motor symptoms of Parkinson's disease further support this hypothesis. Thus, in our collaborative research efforts, we seek to understand the biological basis for exercise-induced protection in order to assist in the development of a safe and clinically effective intervention based on increased physical activity. In addition, we recognize that some individuals cannot or will not engage in physical exercise, and believe that mechanistic studies of exercise-induced protection will provide insights into the development of drugs that could emulate its effects. Using toxins that induce a deficiency of dopamine, we have affirmed that physical exercise can reduce behavioral and neurobiological deficits induced by such toxins, and suggest that these neuroprotective effects are likely to involve the activation of signaling cascades by neurotrophic factors such as glial cell line derived neurotrophic factor.


Dopaminergic Neurons/physiology , Exercise/physiology , Neuroprotective Agents/metabolism , Parkinson Disease/metabolism , Parkinson Disease/therapy , Animals , Dopamine/physiology , Dopaminergic Neurons/metabolism , Humans , Parkinson Disease/physiopathology
18.
PLoS One ; 6(10): e24722, 2011.
Article En | MEDLINE | ID: mdl-22022363

Mild stress can protect against a larger insult, a phenomenon termed preconditioning or tolerance. To determine if a low intensity stressor could also protect cells against intense oxidative stress in a model of dopamine deficiency associated with Parkinson disease, we used methamphetamine to provide a mild, preconditioning stress, 6-hydroxydopamine (6-OHDA) as a source of potentially toxic oxidative stress, and MN9D cells as a model of dopamine neurons. We observed that prior exposure to subtoxic concentrations of methamphetamine protected these cells against 6-OHDA toxicity, whereas higher concentrations of methamphetamine exacerbated it. The protection by methamphetamine was accompanied by decreased uptake of both [(3)H] dopamine and 6-OHDA into the cells, which may have accounted for some of the apparent protection. However, a number of other effects of methamphetamine exposure suggest that the drug also affected basic cellular survival mechanisms. First, although methamphetamine preconditioning decreased basal pERK1/2 and pAkt levels, it enhanced the 6-OHDA-induced increase in these phosphokinases. Second, the apparent increase in pERK1/2 activity was accompanied by increased pMEK1/2 levels and decreased activity of protein phosphatase 2. Third, methamphetamine upregulated the pro-survival protein Bcl-2. Our results suggest that exposure to low concentrations of methamphetamine cause a number of changes in dopamine cells, some of which result in a decrease in their vulnerability to subsequent oxidative stress. These observations may provide insights into the development of new therapies for prevention or treatment of PD.


Cytoprotection/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Methamphetamine/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Cell Death/drug effects , Cell Line , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopaminergic Neurons/enzymology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Methamphetamine/toxicity , Mice , Mice, Inbred C57BL , Neuroprotective Agents/toxicity , Oxidopamine/toxicity , Phosphorylation/drug effects , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Protons , Superoxide Dismutase/metabolism
19.
Prog Neurobiol ; 95(3): 373-95, 2011 Nov.
Article En | MEDLINE | ID: mdl-21930182

Silent information regulator two proteins (sirtuins or SIRTs) are a group of histone deacetylases whose activities are dependent on and regulated by nicotinamide adenine dinucleotide (NAD(+)). They suppress genome-wide transcription, yet upregulate a select set of proteins related to energy metabolism and pro-survival mechanisms, and therefore play a key role in the longevity effects elicited by calorie restriction. Recently, a neuroprotective effect of sirtuins has been reported for both acute and chronic neurological diseases. The focus of this review is to summarize the latest progress regarding the protective effects of sirtuins, with a focus on SIRT1. We first introduce the distribution of sirtuins in the brain and how their expression and activity are regulated. We then highlight their protective effects against common neurological disorders, such as cerebral ischemia, axonal injury, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. Finally, we analyze the mechanisms underlying sirtuin-mediated neuroprotection, centering on their non-histone substrates such as DNA repair enzymes, protein kinases, transcription factors, and coactivators. Collectively, the information compiled here will serve as a comprehensive reference for the actions of sirtuins in the nervous system to date, and will hopefully help to design further experimental research and expand sirtuins as therapeutic targets in the future.


Nervous System Physiological Phenomena , Neuroprotective Agents , Sirtuins/physiology , Animals , Brain Chemistry/genetics , Caloric Restriction , Cell Nucleus/drug effects , Cytosol/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Histone Deacetylases/physiology , Humans , Longevity/physiology , Mitochondria/drug effects , Nervous System Physiological Phenomena/drug effects , Neurodegenerative Diseases/drug therapy , Sirtuin 1/metabolism , Sirtuin 1/physiology , Sirtuins/genetics , Sirtuins/metabolism , Sirtuins/pharmacology , Sirtuins/therapeutic use
20.
Urol Oncol ; 29(1): 100-3, 2011.
Article En | MEDLINE | ID: mdl-21194645

Suggested approaches to reduce the occurrence of plagiarism in academia, particularly among trainees. These include (1) educating individuals as to the definition of plagiarism and its consequences through written guidelines, active discussions, and practice in identifying proper and improper citation practices; (2) distributing checklists that break the writing task into more manageable steps, (3) requiring the submission of an outline and then a first draft prior to the deadline for a paper; (4) making assignments relevant to individual interests; and (5) providing trainees with access to software programs that detect plagiarism.


Biomedical Research/education , Plagiarism , Teaching , Writing , Humans , Software , Students
...