Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Neurotoxicology ; 83: 69-76, 2021 03.
Article in English | MEDLINE | ID: mdl-33400970

ABSTRACT

On the basis of the evidence that extracellular Zn2+ influx induced with AMPA causes Parkinson's syndrome in rats that apomorphine-induced movement disorder emerges, here we used a low dose of AMPA, which does not increase intracellular Zn2+ level in the substantia nigra pars compacta (SNpc) of young adult rats, and tested whether intracellular Zn2+ dysregulation induced with AMPA is accelerated in the SNpc of aged rats, resulting in age-related vulnerability to Parkinson's syndrome. When AMPA (1 mM) was injected at the rate of 0.05 µl/min for 20 min into the SNpc, intracellular Zn2+ level was increased in the SNpc of aged rats followed by increase in turning behavior in response to apomorphine and nigral dopaminergic degeneration. In contrast, young adult rats do not show movement disorder and nigral dopaminergic degeneration, in addition to no increase in intracellular Zn2+. In aged rats, movement disorder and nigral dopaminergic degeneration were rescued by co-injection of either extracellular (CaEDTA) or intracellular (ZnAF-2DA) Zn2+ chelators. 1-Naphthyl acetyl spermine (NASPM), a selective blocker of Ca2+- and Zn2+-permeable GluR2-lacking AMPA receptors blocked increase in intracellular Zn2+ in the SNpc of aged rats followed by rescuing nigral dopaminergic degeneration. The present study indicates that intracellular Zn2+ dysregulation is accelerated by Ca2+- and Zn2+-permeable GluR2-lacking AMPA receptor activation in the SNpc of aged rats, resulting in age-related vulnerability to Parkinson's syndrome.


Subject(s)
Dopaminergic Neurons/drug effects , Excitatory Amino Acid Agonists/toxicity , Nerve Degeneration , Parkinson Disease, Secondary/chemically induced , Pars Compacta/drug effects , Receptors, AMPA/agonists , Zinc/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/toxicity , Age Factors , Animals , Behavior, Animal/drug effects , Calcium/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Male , Motor Activity/drug effects , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Parkinson Disease, Secondary/physiopathology , Pars Compacta/metabolism , Pars Compacta/pathology , Pars Compacta/physiopathology , Rats, Wistar , Receptors, AMPA/metabolism
2.
Neurosci Lett ; 739: 135443, 2020 11 20.
Article in English | MEDLINE | ID: mdl-33141067

ABSTRACT

Parkinson's Disease (PD) patients undergoing subthalamic nucleus deep brain stimulation (STN-DBS) therapy can reduce levodopa equivalent daily dose (LEDD) by approximately 50 %, leading to less symptoms of dyskinesia. The underlying mechanisms contributing to this reduction remain unclear, but studies posit that STN-DBS may increase striatal dopamine levels by exciting remaining dopaminergic cells in the substantia nigra pars compacta (SNc). Yet, no direct evidence has shown how SNc neuronal activity responds during STN-DBS in PD. Here, we use a hemiparkinsonian rat model of PD and employ in vivo electrophysiology to examine the effects of STN-DBS on SNc neuronal spiking activity. We found that 43 % of SNc neurons in naïve rats reduced their spiking frequency to 29.8 ± 18.5 % of baseline (p = 0.010). In hemiparkinsonian rats, a higher number of SNc neurons (88 % of recorded cells) decreased spiking frequency to 61.6 ± 4.4 % of baseline (p = 0.030). We also noted that 43 % of SNc neurons in naïve rats increased spiking frequency from 0.2 ± 0.0 Hz at baseline to 1.8 ± 0.3 Hz during stimulation, but only 1 SNc neuron from 1 hemiparkinsonian rat increased its spiking frequency by 12 % during STN-DBS. Overall, STN-DBS decreased spike frequency in the majority of recorded SNc neurons in a rat model of PD. Less homogenous responsiveness in directionality in SNc neurons during STN-DBS was seen in naive rats. Plausibly, poly-synaptic network signaling from STN-DBS may underlie these changes in SNc spike frequencies.


Subject(s)
Action Potentials , Neurons/physiology , Parkinsonian Disorders/physiopathology , Pars Compacta/physiopathology , Subthalamic Nucleus/physiopathology , Animals , Disease Models, Animal , Electric Stimulation , Male , Parkinson Disease/physiopathology , Rats, Sprague-Dawley
3.
Exp Neurol ; 333: 113433, 2020 11.
Article in English | MEDLINE | ID: mdl-32791155

ABSTRACT

GABAergic neurons in the rostromedial tegmental nucleus (RMTg) receive major input from the lateral habenula (LHb), which conveys negative reward and motivation related information, and project intensively to midbrain dopamine neurons, including those in the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc). The RMTg-VTA circuit has been shown to be linked to the affective behavior, but the role of the RMTg-SNc circuit in aversion and depression has not been well understood. This study demonstrated that exciting or inhibiting VgatRMTg-SNc neurons was sufficient to increase or decrease immobility time in the forced swim test (FST), respectively. Furthermore, exciting the VgatRMTg-SNc pathway caused aversive behavior. Ninety percent of the SNc putative dopamine neurons were inhibited in extracellular recordings. Furthermore, inhibiting the VgatRMTg-SNc pathway reversed behavioral despair in chronic restraint stress (CRS) depression model mice. Manipulations of the pathway did not affect the hedonic value of the reward in the sucrose-preference test (SPT) or general motor function. In conclusion, these results indicate that the VgatRMTg-SNc pathway regulates aversive and despair behavior, which suggests that the RMTg may mediate the role of LHb in negative behaviors through regulating the activity of SNc neurons.


Subject(s)
Avoidance Learning , Pars Compacta/physiopathology , Stress, Psychological/psychology , Ventral Tegmental Area/physiopathology , Anhedonia , Animals , Depression/physiopathology , Depression/psychology , Dopaminergic Neurons , Female , Male , Mice , Motor Activity , Neurons , Restraint, Physical , Reward , Signal Transduction , Swimming/psychology , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
4.
J Chem Neuroanat ; 106: 101789, 2020 07.
Article in English | MEDLINE | ID: mdl-32334030

ABSTRACT

This study examined the effect of prolonged water deprivation, in rat, on 5-HT and TH- immuno-expression in Dorsal Raphe Nucleus (DRN), Substantia Nigra pars compacta (SNc), Ventral Tegmental Area (VTA), and Magnus Raphe Nucleus (MRN). In parallel, we evaluated the anxiety state and pain perception in dehydrated rats. Our Findings revealed that dehydrated rats exhibited more preference for the dark compartment, suggesting that prolonged water deprivation is associated to an anxiogenic effect. After one week, 5 H T IR in the DRN of dehydrated rates showed a significant decrease. This was reversed to a significant increase post week 2 of dehydration. Our findings also demonstrated that TH-IR in DRN, MRN, SNc and VTA neuronal systems is significantly and gradually enhanced after 1-and-2-week osmotic stress. In addition, our results proved that all dehydrated rats were characterized by a significant and proportional rise of the reaction time to the nociceptive response in the hot plate test, as water deprivation duration increased, suggesting that dehydration caused a significant decrease in pain perception. Finally, the data described here clearly showed the implication of serotonin and dopamine neurotransmitter systems in the resistance to osmotic stress. Therefore, in this study, such central impairments were traduced by a few peripheral outcomes manifested by changes in mood state and nociception.


Subject(s)
Anxiety/metabolism , Pain Perception/physiology , Serotonin/metabolism , Tyrosine 3-Monooxygenase/metabolism , Water Deprivation/physiology , Animals , Anxiety/physiopathology , Behavior, Animal/physiology , Dorsal Raphe Nucleus/metabolism , Dorsal Raphe Nucleus/physiopathology , Male , Nucleus Raphe Magnus/metabolism , Nucleus Raphe Magnus/physiopathology , Osmotic Pressure/physiology , Pars Compacta/metabolism , Pars Compacta/physiopathology , Rats , Rats, Wistar , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/physiopathology
5.
Mol Pharmacol ; 98(6): 730-738, 2020 12.
Article in English | MEDLINE | ID: mdl-32015009

ABSTRACT

Parkinson disease (PD) is a devastating, largely nonfamilial, age-related disorder caused by the progressive loss of dopamine (DA) neurons in the substantia nigra pars compacta (SNc). Release of DA from these neurons into the dorsal striatum is crucial for regulating movement and their loss causes PD. Unfortunately, the mechanisms underlying SNc neurodegeneration remain unclear, and currently there is no cure for PD, only symptomatic treatments. Recently, several regulator of G protein signaling (RGS) proteins have emerged as critical modulators of PD pathogenesis and/or motor dysfunction and dyskinesia: RGSs 4, 6, 9, and 10. Striatal RGS4 has been shown to exacerbate motor symptoms of DA loss by suppressing M4-autoreceptor-Gα i/o signaling in striatal cholinergic interneurons. RGS6 and RGS9 are key regulators of D2R-Gα i/o signaling in SNc DA neurons and striatal medium spiny neurons, respectively. RGS6, expressed in human and mouse SNc DA neurons, suppresses characteristic PD hallmarks in aged mice, including SNc DA neuron loss, motor deficits, and α-synuclein accumulation. After DA depletion, RGS9 (through its inhibition of medium spiny neuron D2R signaling) suppresses motor dysfunction induced by L-DOPA or D2R-selective agonists. RGS10 is highly expressed in microglia, the brain's resident immune cells. Within the SNc, RGS10 may promote DA neuron survival through the upregulation of prosurvival genes and inhibition of microglial inflammatory factor expression. Thus, RGSs 4, 6, 9, and 10 are critical modulators of cell signaling pathways that promote SNc DA neuron survival and/or proper motor control. Accordingly, these RGS proteins represent novel therapeutic targets for the treatment of PD pathology. SIGNIFICANCE STATEMENT: Parkinson disease (PD), the most common movement disorder, is a progressive neurodegenerative disease characterized by substantia nigra pars compacta (SNc) dopamine (DA) neuron loss and subsequent motor deficits. Current PD therapies only target disease motor symptomology and are fraught with side effects. Therefore, researchers have begun to explore alternative therapeutic options. Regulator of G protein signaling (RGS) proteins, whether primarily expressed in SNc DA neurons (RGS6), striatal neurons (RGSs 4 and 9), or microglia (RGS10), modulate key signaling pathways important for SNc DA neuron survival and/or proper motor control. As such, RGS proteins represent novel therapeutic targets in PD.


Subject(s)
Motor Activity/physiology , Nerve Degeneration/pathology , Parkinson Disease/pathology , Pars Compacta/pathology , RGS Proteins/metabolism , Animals , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , GTP-Binding Proteins/metabolism , Humans , Mice , Microglia/metabolism , Parkinson Disease/physiopathology , Pars Compacta/physiopathology , Signal Transduction/physiology
6.
Braz J Med Biol Res ; 52(7): e8303, 2019.
Article in English | MEDLINE | ID: mdl-31291382

ABSTRACT

Reinforcement omission effects (ROEs) are characterized by higher response rates after reinforcement omission than after reinforcement delivery. This pattern of behavior is interpreted in terms of motivational and attentional processes. Recent studies from our laboratory have shown that the amygdala, nucleus accumbens, and medial prefrontal cortex are involved in ROE modulation. Also, the literature has demonstrated a role of other areas such as substantia nigra pars compacta (SNc) and the ventral tegmental area (VTA) in processes related to surprising events, such as prediction error and presentation or omission of an event (exteroceptive stimulus and reinforcement). Since these structures send projections to areas related to ROE modulation such as the amygdala, nucleus accumbens, and prefrontal cortex, the objective of the present study was to determine whether the SNc and VTA also integrate the circuit involved in ROE modulation. Rats were trained on a fixed-interval 12 s with limited-hold 6 s signaled schedule of reinforcement (Pre-lesion training). After acquisition of stable performance, the rats received bilateral neurotoxic lesions of the SNc (Experiment 1) and VTA (Experiment 2). Following postoperative recovery, the rats were submitted to two refresher sessions (Post-lesion training). Subsequently, the training was changed from a 100 to a 50% schedule of reinforcement (Post-lesion testing). In both experiments, the results showed that there was no difference in performance between sham rats and rats with bilateral lesions of the SNc or the VTA.


Subject(s)
Behavior, Animal/physiology , Conditioning, Operant/physiology , Pars Compacta/injuries , Reinforcement, Psychology , Substantia Nigra/injuries , Ventral Tegmental Area/injuries , Animals , Learning/physiology , Male , Pars Compacta/physiopathology , Rats , Rats, Wistar , Substantia Nigra/physiopathology , Ventral Tegmental Area/physiopathology
7.
Mov Disord ; 34(8): 1130-1143, 2019 08.
Article in English | MEDLINE | ID: mdl-31216379

ABSTRACT

Studies of the pathophysiology of parkinsonism (specifically akinesia and bradykinesia) have a long history and primarily model the consequences of dopamine loss in the basal ganglia on the function of the basal ganglia/thalamocortical circuit(s). Changes of firing rates of individual nodes within these circuits were originally considered central to parkinsonism. However, this view has now given way to the belief that changes in firing patterns within the basal ganglia and related nuclei are more important, including the emergence of burst discharges, greater synchrony of firing between neighboring neurons, oscillatory activity patterns, and the excessive coupling of oscillatory activities at different frequencies. Primarily focusing on studies obtained in nonhuman primates and human patients with Parkinson's disease, this review summarizes the current state of this field and highlights several emerging areas of research, including studies of the impact of the heterogeneity of external pallidal neurons on parkinsonism, the importance of extrastriatal dopamine loss, parkinsonism-associated synaptic and morphologic plasticity, and the potential role(s) of the cerebellum and brainstem in the motor dysfunction of Parkinson's disease. © 2019 International Parkinson and Movement Disorder Society.


Subject(s)
Basal Ganglia/physiopathology , Cerebral Cortex/physiopathology , Dopamine/metabolism , Parkinson Disease/physiopathology , Thalamus/physiopathology , Animals , Basal Ganglia/metabolism , Brain Stem/metabolism , Brain Stem/physiopathology , Brain Waves/physiology , Cerebellum/metabolism , Cerebellum/physiopathology , Cerebral Cortex/metabolism , Electroencephalography , Globus Pallidus/metabolism , Globus Pallidus/physiopathology , Haplorhini , Humans , Neostriatum/metabolism , Neostriatum/physiopathology , Neural Pathways/metabolism , Neural Pathways/physiopathology , Neuronal Plasticity , Parkinson Disease/metabolism , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/physiopathology , Pars Compacta/metabolism , Pars Compacta/physiopathology , Thalamus/metabolism
8.
Neural Plast ; 2019: 8215017, 2019.
Article in English | MEDLINE | ID: mdl-31073303

ABSTRACT

Physical training confers protection to dopaminergic neurons in rodent models of parkinsonism produced by neurotoxins. The sparing effect of physical training on dopaminergic neurons can be tested with training applied during chronic MPTP treatment, while the neurorestorative effect when training is applied after completing such treatment. In this study, the effect of the onset of training respective to chronic MPTP treatment was specifically addressed. Three groups of mice were injected with 10 doses of MPTP (12.5 mg/kg/injection) over 5 weeks. The first group remained sedentary; the second one underwent early onset training, which started 1 week before commencing MPTP treatment, continued throughout 5 weeks of treatment and 4 weeks thereafter; the third group underwent late-onset training of the same length and intensity as the former group, except that it started immediately after the end of MPTP treatment. Two groups served as controls: a saline-injected group that remained sedentary and saline-injected group, which underwent the same training as the early and late-onset training groups. Both early and late-onset physical training saved almost all nigral and VTA dopaminergic neurons, prevented inflammatory response, and increased the BDNF and GDNF levels to a similar extent. From these results one may conclude that early and late-onset training schedules were equipotent in their neuroprotective effect and that the mechanism of neuroprotection was similar. The sparing effect of early onset training may be satisfactorily explained by assuming that the increased level of BDNF and GDNF prevented the degeneration of dopaminergic neurons. To explain a similar number of dopaminergic neurons detected at the end of the early and late-onset training, one should additionally assume that the former training schedule induced neurogenesis. Results of this study support the view that physical activity may be neuroprotective even at a more advanced stage of PD and justify starting physical activity at any point of the disease.


Subject(s)
Dopaminergic Neurons/physiology , Exercise Therapy , Neuronal Plasticity , Parkinson Disease/prevention & control , Parkinson Disease/physiopathology , Animals , Astrocytes/physiology , Chronic Disease/prevention & control , Disease Models, Animal , Male , Mice, Inbred C57BL , Microglia/physiology , Parkinsonian Disorders/physiopathology , Parkinsonian Disorders/prevention & control , Pars Compacta/physiopathology , Ventral Tegmental Area/physiopathology
9.
Glia ; 67(8): 1542-1557, 2019 08.
Article in English | MEDLINE | ID: mdl-31025779

ABSTRACT

Parkinson's disease (PD) is characterized by the selective degeneration of dopamine (DA) neurons of the substantia nigra pars compacta (SN), while the neighboring ventral tegmental area (VTA) is relatively spared. The mechanisms underlying this selectivity are not fully understood. Here, we demonstrate a vital role for subregional astrocytes in the protection of VTA DA neurons. We found that elimination of astrocytes in vitro exposes a novel vulnerability of presumably protected VTA DA neurons to the PD mimetic toxin MPP+ , as well as exacerbation of SN DA neuron vulnerability. Conversely, VTA astrocytes protected both VTA and SN DA neurons from MPP+ toxicity in a dose dependent manner, and this protection was mediated via a secreted molecule. RNAseq analysis of isolated VTA and SN astrocytes demonstrated a vast array of transcriptional differences between these two closely related populations demonstrating regional heterogeneity of midbrain astrocytes. We found that GDF15, a member of the TGFß superfamily which is expressed 230-fold higher in VTA astrocytes than SN, recapitulates neuroprotection of both rat midbrain and iPSC-derived DA neurons, whereas its knockdown conversely diminished this effect. Neuroprotection was likely mediated through the GRFAL receptor expressed on DA neurons. Together; these results suggest that subregional differences in astrocytes underlie the selective degeneration or protection of DA neurons in PD.


Subject(s)
Astrocytes/physiology , Nerve Degeneration/physiopathology , Neuroprotection/physiology , Parkinsonian Disorders/physiopathology , Animals , Cells, Cultured , Coculture Techniques , Dopaminergic Neurons/physiology , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Growth Differentiation Factor 15/metabolism , Humans , Induced Pluripotent Stem Cells/physiology , Pars Compacta/physiopathology , Rats, Transgenic , Ventral Tegmental Area/physiopathology
10.
Braz. j. med. biol. res ; 52(7): e8303, 2019. graf
Article in English | LILACS | ID: biblio-1011594

ABSTRACT

Reinforcement omission effects (ROEs) are characterized by higher response rates after reinforcement omission than after reinforcement delivery. This pattern of behavior is interpreted in terms of motivational and attentional processes. Recent studies from our laboratory have shown that the amygdala, nucleus accumbens, and medial prefrontal cortex are involved in ROE modulation. Also, the literature has demonstrated a role of other areas such as substantia nigra pars compacta (SNc) and the ventral tegmental area (VTA) in processes related to surprising events, such as prediction error and presentation or omission of an event (exteroceptive stimulus and reinforcement). Since these structures send projections to areas related to ROE modulation such as the amygdala, nucleus accumbens, and prefrontal cortex, the objective of the present study was to determine whether the SNc and VTA also integrate the circuit involved in ROE modulation. Rats were trained on a fixed-interval 12 s with limited-hold 6 s signaled schedule of reinforcement (Pre-lesion training). After acquisition of stable performance, the rats received bilateral neurotoxic lesions of the SNc (Experiment 1) and VTA (Experiment 2). Following postoperative recovery, the rats were submitted to two refresher sessions (Post-lesion training). Subsequently, the training was changed from a 100 to a 50% schedule of reinforcement (Post-lesion testing). In both experiments, the results showed that there was no difference in performance between sham rats and rats with bilateral lesions of the SNc or the VTA.


Subject(s)
Animals , Male , Rats , Reinforcement, Psychology , Behavior, Animal/physiology , Substantia Nigra/injuries , Ventral Tegmental Area/injuries , Conditioning, Operant/physiology , Pars Compacta/injuries , Substantia Nigra/physiopathology , Rats, Wistar , Ventral Tegmental Area/physiopathology , Pars Compacta/physiopathology , Learning/physiology
12.
Neurotox Res ; 34(4): 799-807, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29611150

ABSTRACT

Non-motor symptoms are increasingly identified to present clinical and diagnostic importance for Parkinson's disease (PD). The multifactorial origin of pain in PD makes this symptom of great complexity. The dopamine precursor, L-DOPA (L-3,4-dihydroxyphenylalanine), the classic therapy for PD, seems to be effective in pain threshold; however, there are no studies correlating L-DOPA-induced dyskinesia (LID) and nociception development in experimental Parkinsonism. Here, we first investigated nociceptive responses in a 6-hydroxydopamine (6-OHDA)-lesioned rat model of Parkinson's disease to a hind paw-induced persistent inflammation. Further, the effect of L-DOPA on nociception behavior at different times of treatment was investigated. Pain threshold was determined using von Frey and Hot Plate/Tail Flick tests. Dyskinesia was measured by abnormal involuntary movements (AIMs) induced by L-DOPA administration. This data is consistent to show that 6-OHDA-lesioned rats had reduced nociceptive thresholds compared to non-lesioned rats. Additionally, when these rats were exposed to a persistent inflammatory challenge, we observed increased hypernociceptive responses, namely hyperalgesia. L-DOPA treatment alleviated pain responses on days 1 and 7 of treatment, but not on day 15. During that period, we observed an inverse relationship between LID and nociception threshold in these rats, with a high LID rate corresponding to a reduced nociception threshold. Interestingly, pain responses resulting from CFA-induced inflammation were significantly enhanced during established dyskinesia. These data suggest a pro-algesic effect of L-DOPA-induced dyskinesia, which is confirmed by the correlation founded here between AIMs and nociceptive indexes. In conclusion, our results are consistent with the notion that central dopaminergic mechanism is directly involved in nociceptive responses in Parkinsonism condition.


Subject(s)
Antiparkinson Agents/toxicity , Dyskinesia, Drug-Induced/physiopathology , Levodopa/toxicity , Nociceptive Pain/physiopathology , Parkinsonian Disorders/drug therapy , Animals , Antiparkinson Agents/pharmacology , Corpus Striatum/physiopathology , Inflammation/physiopathology , Levodopa/pharmacology , Male , Oxidopamine , Parkinsonian Disorders/physiopathology , Pars Compacta/physiopathology , Rats, Wistar
13.
J Biomed Sci ; 25(1): 26, 2018 Mar 19.
Article in English | MEDLINE | ID: mdl-29554962

ABSTRACT

BACKGROUND: Parkinson's disease is described as resulting from dopaminergic cells progressive degeneration, specifically in the substantia nigra pars compacta that influence the voluntary movements control, decision making and time perception. AIM: This review had a goal to update the relation between time perception and Parkinson's Disease. METHODOLOGY: We used the PRISMA methodology for this investigation built guided for subjects dopaminergic dysfunction in the time judgment, pharmacological models with levodopa and new studies on the time perception in Parkinson's Disease. We researched on databases Scielo, Pubmed / Medline and ISI Web of Knowledge on August 2017 and repeated in September 2017 and February 2018 using terms and associations relevant for obtaining articles in English about the aspects neurobiology incorporated in time perception. No publication status or restriction of publication date was imposed, but we used as exclusion criteria: dissertations, book reviews, conferences or editorial work. RESULTS/DISCUSSION: We have demonstrated that the time cognitive processes are underlying to performance in cognitive tasks and that many are the brain areas and functions involved and the modulators in the time perception performance. CONCLUSIONS: The influence of dopaminergic on Parkinson's Disease is an important research tool in Neuroscience while allowing for the search for clarifications regarding behavioral phenotypes of Parkinson's disease patients and to study the areas of the brain that are involved in the dopaminergic circuit and their integration with the time perception mechanisms.


Subject(s)
Basal Ganglia/physiopathology , Parkinson Disease/physiopathology , Pars Compacta/physiopathology , Time Perception , Basal Ganglia/chemistry , Pars Compacta/chemistry , Synaptic Transmission/physiology
14.
Neuroscience ; 379: 292-301, 2018 05 21.
Article in English | MEDLINE | ID: mdl-29559387

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder caused by loss of dopaminergic neurons in the substantia nigra, leading to motor dysfunction. Growing evidence has demonstrated that endurance exercise (EE) confers neuroprotection against PD. However, the exact molecular mechanisms responsible for exercise-induced protection of dopaminergic neurons in PD remain unclear. Since oxidative stress plays a key role in the degenerative process of PD. We investigated whether EE-induced neuroprotection is associated with enhanced antioxidative capacity and autophagy, using a mouse model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration. C57BL/6 male mice were randomly assigned to four groups: control (CON, n = 12), exercise (EXE, n = 12), MPTP (MPTP, n = 12) and MPTP + exercise (MPTP + EXE, n = 12). Our data demonstrated that while MPTP treatment impaired motor function, EE restored MPTP-induced motor deficits. Our biochemical data showed that EE-induced neuroprotection occurs in combination with multiple synergic neuroprotective pathways: (1) increased neurogenesis shown by an increase in BrdU-positive neurons; (2) diminished loss of dopaminergic neurons evidenced by upregulated tyrosine hydroxylase (TH) and dopamine transporter (DAT) levels; (3) increased antioxidant capacity (e.g., CuZnSOD, CATALASE, GPX1/2, HO-1, DJ1 and PRXIII); and (4) enhanced autophagy (LC3 II, p62, BECLIN1, BNIP3, LAMP2, CATHEPSIN L and TFEB). Our study suggests that EE-induced multiple synergic protective pathways including enhanced neurogenesis, antioxidative capacity, and concordant autophagy promotion contribute to restoration of impaired dopaminergic neuronal function caused by PD. Thus, PD patients should be encouraged to actively participate in regular EE as a potent nonpharmacological therapeutic strategy against PD.


Subject(s)
Antioxidants/metabolism , Autophagy/physiology , Endurance Training , MPTP Poisoning/therapy , Neurogenesis/physiology , Neuroprotection/physiology , Animals , Dopaminergic Neurons/pathology , Dopaminergic Neurons/physiology , Hippocampus/pathology , Hippocampus/physiopathology , MPTP Poisoning/pathology , MPTP Poisoning/physiopathology , Male , Mice, Inbred C57BL , Pars Compacta/pathology , Pars Compacta/physiopathology , Random Allocation
15.
Behav Brain Res ; 344: 42-47, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29452192

ABSTRACT

Recent studies have established methods for establishing a rodent model that mimics progressive stages of human Parkinson's disease (PD), via injection of graded doses of 6-hydroxydopamine (6-OHDA) into regions within the nigrostriatal pathway. However, the electrophysiological characteristics of the subthalamic nucleus (STN) in this model have not been fully elucidated in this model. This study aimed to investigate changes in the neuronal activity of the STN in a graded mouse model of PD. Increasing doses of 6-OHDA were unilaterally injected into the medial forebrain bundle (MFB) to produce a hemi-parkinsonian mouse model, mimicking early, moderate, advanced, and severe stages of human PD. Mice treated with higher doses of 6-OHDA demonstrated significantly lower rates of use of the impaired (contralateral) forelimb during wall contact, relative to sham mice. The STN firing rate was significantly increased in groups with >75% dopaminergic cell loss in the substantia nigra pars compacta (SNc), whereas little increase was observed in groups with partial lesions of the SNc, relative to the sham group. In addition, firing patterns of the STN in groups treated with higher doses of 6-OHDA became more irregular and exhibited burst-like patterns of activity, with dominant slow wave oscillations in the frequency range of 0.3-2.5 Hz. Our results demonstrated a strong correlation between neuronal activities in the STN and dopamine depletion in the nigrostriatal pathway, which can be manipulated by variation of 6-OHDA doses.


Subject(s)
Action Potentials/physiology , Dopamine/deficiency , Dopaminergic Neurons/physiology , Parkinsonian Disorders/physiopathology , Pars Compacta/physiopathology , Subthalamic Nucleus/physiopathology , Animals , Cell Death , Disease Progression , Dopaminergic Neurons/pathology , Dose-Response Relationship, Drug , Forelimb/physiopathology , Functional Laterality , Medial Forebrain Bundle/drug effects , Medial Forebrain Bundle/pathology , Medial Forebrain Bundle/physiopathology , Mice, Inbred C57BL , Motor Activity/physiology , Neural Pathways/pathology , Neural Pathways/physiopathology , Oxidopamine/toxicity , Parkinsonian Disorders/pathology , Pars Compacta/pathology , Random Allocation , Subthalamic Nucleus/pathology , Tyrosine 3-Monooxygenase/metabolism
16.
Biochem Biophys Res Commun ; 496(4): 1102-1108, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29378186

ABSTRACT

Ghrelin plays roles in a wide range of central functions by activating the growth hormone secretagogue receptor (GHSR). This receptor has recently been found in the substantia nigra (SN) to control dopamine (DA)-related physiological functions. The dysregulation of DA neurons in the SN pars compacta (SNc) and the consequent depletion of striatal DA are known to underlie the motor deficits observed in Parkinson's disease (PD). In the present study, we further investigated the role of the SN-ghrelin system in motor function under the stereotaxic injection of AAV-CMV-FLEX-diphtheria toxin A (DTA) into the SN of dopamine transporter (DAT)-Cre (DATSN::DTA) mice to expunge DA neurons of the SNc. First, we confirmed the dominant expression of GHSR1a, which is a functional GHSR, in tyrosine hydroxylase (TH)-positive DA neurons in the SNc of control mice. In DATSN::DTA mice, we clearly observed motor dysfunction using several behavioral tests. An immunohistochemical study revealed a dramatic loss of TH-positive DA neurons in the SNc and DAT-labeled axon terminals in the striatum, and an absence of mRNAs for TH and DAT in the SN of DATSN::DTA mice. The mRNA level of GHSR1a was drastically decreased in the SN of these mice. In normal mice, we also found the mRNA expression of GHSR1a within GABAergic neurons in the SN pars reticulata (SNr). Under these conditions, a single injection of ghrelin into the SN failed to improve the motor deficits caused by ablation of the nigrostriatal DA network using DATSN::DTA mice, whereas intra-SN injection of ghrelin suppressed the motor dysfunction caused by the administration of haloperidol, which is associated with the transient inhibition of DA transmission. These findings suggest that phasic activation of the SNc-ghrelin system could improve the dysregulation of nigrostriatal DA transmission related to the initial stage of PD, but not the motor deficits under the depletion of nigrostriatal DA. Although GHSRs are found in non-DA cells of the SNr, GHSRs on DA neurons in the SNc may play a crucial role in motor function.


Subject(s)
Dopaminergic Neurons/metabolism , Ghrelin/administration & dosage , Movement Disorders/drug therapy , Movement Disorders/physiopathology , Pars Compacta/physiopathology , Receptors, Dopamine/metabolism , Ablation Techniques , Animals , Dopamine Antagonists/administration & dosage , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Dose-Response Relationship, Drug , Male , Mice , Mice, Transgenic , Neurosurgical Procedures , Pars Compacta/drug effects , Recovery of Function/drug effects , Treatment Outcome
17.
Gastroenterology ; 153(6): 1581-1593, 2017 12.
Article in English | MEDLINE | ID: mdl-28912019

ABSTRACT

BACKGROUND & AIMS: In most patients with Parkinson's disease, gastrointestinal (GI) dysfunctions, such as gastroparesis and constipation, are prodromal to the cardinal motor symptoms of the disease. Sporadic Parkinson's disease has been proposed to develop after ingestion of neurotoxicants that affect the brain-gut axis via the vagus nerve, and then travel to higher centers, compromising the substantia nigra pars compacta (SNpc) and, later, the cerebral cortex. We aimed to identify the pathway that connects the brainstem vagal nuclei and the SNpc, and to determine whether this pathway is compromised in a rat model of Parkinsonism. METHODS: To study this neural pathway in rats, we placed tracers in the dorsal vagal complex or SNpc; brainstem and midbrain were examined for tracer distribution and neuronal neurochemical phenotype. Rats were given injections of paraquat once weekly for 3 weeks to induce features of Parkinsonism, or vehicle (control). Gastric tone and motility were recorded after N-methyl-d-aspartate microinjection in the SNpc and/or optogenetic stimulation of nigro-vagal terminals in the dorsal vagal complex. RESULTS: Stimulation of the SNpc increased gastric tone and motility via activation of dopamine 1 receptors in the dorsal vagal complex. In the paraquat-induced model of Parkinsonism, this nigro-vagal pathway was compromised during the early stages of motor deficit development. CONCLUSIONS: We identified and characterized a nigro-vagal monosynaptic pathway in rats that controls gastric tone and motility. This pathway might be involved in the prodromal gastric dysmotility observed in patients with early-stage Parkinson's disease.


Subject(s)
Brain Stem/physiopathology , Gastric Emptying , Parkinson Disease, Secondary/physiopathology , Pars Compacta/physiopathology , Stomach/innervation , Vagus Nerve/physiopathology , Animals , Brain Stem/drug effects , Brain Stem/metabolism , Disease Models, Animal , Gastric Emptying/drug effects , Immunohistochemistry , Male , Motor Activity , Neural Pathways/metabolism , Neural Pathways/physiopathology , Neuroanatomical Tract-Tracing Techniques , Neurotransmitter Agents/pharmacology , Optogenetics , Paraquat , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Pars Compacta/drug effects , Pars Compacta/metabolism , Rats, Sprague-Dawley , Receptors, Dopamine D1/metabolism , Time Factors , Vagus Nerve/drug effects , Vagus Nerve/metabolism
18.
Neuroscience ; 348: 83-97, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28212832

ABSTRACT

Pedunculopontine nucleus (PPN) has been considered a critically important region in the regulation of some of the physiological functions that fail during the progression of Parkinson's disease (PD). In this paper, the effects of unilateral neurotoxic lesion of the PPN [through the injection of N-methyl-d-aspartate (NMDA) solution (concentration: 0.1M; volume: 0.5µL)] in motor execution and gait disorders and the changes in cellular and molecular indicators in rat nigral tissue were evaluated. The motor execution was assessed using the beam test (BT) and the gait disorders by footprint test. Glutathione (GSH) concentrations, acetyl cholinesterase enzymatic activity (AChE EA), and brain-derived neurotrophic factor (BDNF) mRNA expression in nigral tissue were analyzed. NMDA-lesioned rats showed fine motor dysfunction with a significant increase in the slow (p≤0.01) and fast movement (p≤0.01) time and in path deviation (p≤0.01) on the smaller diameter beams. Moreover, NMDA-lesioned rats exhibited an imprecise path with moments of advances and setbacks, alternating with left and right deviations, suspensions, and inverted positions. Footprint test revealed slight gait disorders, which were manifested by a reduction in the left and right stride lengths, the intra-step distance, and the support area (p≤0.01). Biochemical studies showed that 48h after the PPN neurotoxic injury, the GSH concentrations and BDNF expression were significantly increased (p≤0.01). These variables returned to normal values 7days after the PPN lesion; the AChE EA showed a significant increase at this time. These functional changes in nigral tissue could be a plastic responses associated with early PD.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cholinesterases/metabolism , Gait/physiology , Glutathione/metabolism , Pars Compacta/metabolism , Pedunculopontine Tegmental Nucleus/physiopathology , Animals , Gait/drug effects , Male , Motor Activity/drug effects , Motor Activity/physiology , N-Methylaspartate/toxicity , Pars Compacta/physiopathology , Pedunculopontine Tegmental Nucleus/drug effects , Rats , Rats, Wistar
19.
ACS Chem Neurosci ; 8(4): 702-711, 2017 Apr 19.
Article in English | MEDLINE | ID: mdl-28233992

ABSTRACT

In 1967, L-dopa was introduced as part of the pharmacological therapy of Parkinson's disease (PD) and, in spite of extensive research, no additional effective drugs have been discovered to treat PD. This brings forward the question: why have no new drugs been developed? We consider that one of the problems preventing the discovery of new drugs is that we still have no information on the pathophysiology of the neurodegeneration of the neuromelanin-containing nigrostriatal dopaminergic neurons. Currently, it is widely accepted that the degeneration of dopaminergic neurons, i.e., in the substantia nigra pars compacta, involves mitochondrial dysfunction, the formation of neurotoxic oligomers of alpha-synuclein, the dysfunction of protein degradation systems, neuroinflammation, and oxidative and endoplasmic reticulum stress. However, the initial trigger of these mechanisms in the nigrostriatal system is still unknown. It has been reported that aminochrome induces the majority of these mechanisms involved in the neurodegeneration process. Aminochrome is formed within the cytoplasm of neuromelanin-containing dopaminergic neurons during the oxidation of dopamine to neuromelanin. The oxidation of dopamine to neuromelanin is a normal and harmless process, because healthy individuals have intact neuromelanin-containing dopaminergic neurons. Interestingly, aminochrome-induced neurotoxicity is prevented by two enzymes: DT-diaphorase and glutathione transferase M2-2, which explains why melanin-containing dopaminergic neurons are intact in healthy human brains.


Subject(s)
Dopamine/metabolism , Dopaminergic Neurons/pathology , Nerve Degeneration/metabolism , Parkinson Disease/pathology , Pars Compacta/pathology , Animals , Dopaminergic Neurons/metabolism , Humans , Melanins/metabolism , Nerve Degeneration/physiopathology , Oxidation-Reduction , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Pars Compacta/metabolism , Pars Compacta/physiopathology
20.
Neuroscience ; 340: 359-372, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27826106

ABSTRACT

Anoxic depolarization (AD) is an acute event evoked by brain ischemia, involving a profound loss of cell membrane potential and swelling that spreads over susceptible parts of the gray matter. Its occurrence is a strong predictor of the severity of neuronal injury. Little is known about this event in the Substantia Nigra, a midbrain nucleus critical for motor control. We tested the effects of oxygen and glucose deprivation (OGD), an in vitro model of brain ischemia, in rat midbrain slices. AD developed within 4min from OGD onset and spread in the Substantia Nigra pars reticulata (SNr), but not through the Substantia Nigra pars compacta (SNc). This differential effect involved a contrasting pattern of changes in membrane potential between dopamine-producing SNc and non-dopaminergic SNr neurons. A fast depolarization in SNr neurons was not followed by repolarization after the end of OGD, and was associated with swollen somata and beaded dendrites. In contrast, slowly developing depolarization of SNc neurons led to repolarization after OGD ended, and no changes in neuronal morphology were observed. The AD-resistance of the SNc involved smaller dysregulations of K+ and Ca2+ ions, and a slower loss of energy metabolites. Our results show that acute ischemia profoundly impairs the function and morphology of SNr neurons but not adjacent SNc neurons, and that the surprising higher tolerance of SNc neurons correlates with the resistance of the SNc region to AD. This differential response may affect the pattern of early neuronal injury that develops in the brainstem after acute ischemic insults.


Subject(s)
Brain Ischemia/physiopathology , Neurons/physiology , Pars Compacta/physiopathology , Substantia Nigra/physiopathology , Animals , Brain Ischemia/pathology , Calcium/metabolism , Cations/metabolism , Cell Survival/physiology , Dopamine/metabolism , Female , Glucose/deficiency , Male , Membrane Potentials/physiology , Neurons/pathology , Oxygen/metabolism , Pars Compacta/pathology , Potassium/metabolism , Potassium Channels/metabolism , Rats, Wistar , Substantia Nigra/pathology , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...