Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 125
1.
Methods Mol Biol ; 2761: 491-498, 2024.
Article En | MEDLINE | ID: mdl-38427257

Robust preclinical models of Parkinson's disease (PD) are valuable tools for understanding the biology and treatment of this complex disease. 6-Hydroxydopamine (6-OHDA) is a selective catecholaminergic drug injected into the substantia nigra pars compacta (SNc), medial forebrain bundle (MFB), or striatum, which is then metabolized to induce parkinsonism. Unilateral injection of 6-OHDA produces loss of dopaminergic (DAergic) neurons on the injected side with a marked motor asymmetry known as hemiparkinsonism, typically characterized by a rotational behavior to the impaired side. The present work describes a stable unilateral 6-OHDA-lesioned rat model of PD. 6-OHDA was administered into the MFB, leading to the consistent loss of striatal dopamine (DA) and behavioral imbalance in unilateral 6-OHDA-lesioned rats to establish the model of PD. This model of PD is a valuable tool for understanding the mechanisms underlying the generation of parkinsonian symptoms.


Parkinson Disease , Rats , Male , Animals , Parkinson Disease/metabolism , Oxidopamine/pharmacology , Rats, Wistar , Dopamine/metabolism , Medial Forebrain Bundle/metabolism , Corpus Striatum/metabolism , Substantia Nigra/metabolism , Disease Models, Animal
2.
Sci Rep ; 14(1): 3721, 2024 02 14.
Article En | MEDLINE | ID: mdl-38355892

Parkinson's disease (PD) is the second most common age-related neurodegenerative disease, with a progressive loss of dopaminergic cells and fibers. The purpose of this study was to use different doses of 6-hydroxydopamine (6-OHDA) injection into the medial forebrain bundle (MFB) of mice to mimic the different stages of the disease and to characterize in detail their motor and non-motor behavior, as well as neuropathological features in the nigrostriatal pathway. MFB were injected with 0.5 µg, 1 µg, 2 µg of 6-OHDA using a brain stereotaxic technique. 6-OHDA induced mitochondrial damage dose-dependently, as well as substantia nigra pars compacta (SNpc) tyrosine hydroxylase-positive (TH+) cell loss and striatal TH fiber loss. Activation of astrocytes and microglia in the SNpc and striatum were consistently observed at 7 weeks, suggesting a long-term glial response in the nigrostriatal system. Even with a partial or complete denervation of the nigrostriatal pathway, 6-OHDA did not cause anxiety, although depression-like behavior appeared. Certain gait disturbances were observed in 0.5 µg 6-OHDA lesioned mice, and more extensive in 1 µg group. Despite the loss of more neurons from 2 µg 6-OHDA, there was no further impairment in behaviors compared to 1 µg 6-OHDA. Our data have implications that 1 µg 6-OHDA was necessary and sufficient to induce motor and non-motor symptoms in mice, thus a valuable mouse tool to explore disease progression and new treatment in PD.


Neurodegenerative Diseases , Parkinson Disease , Mice , Animals , Oxidopamine/metabolism , Medial Forebrain Bundle/metabolism , Medial Forebrain Bundle/pathology , Neurodegenerative Diseases/metabolism , Parkinson Disease/metabolism , Dopaminergic Neurons/metabolism , Substantia Nigra/metabolism , Corpus Striatum/metabolism , Disease Models, Animal , Tyrosine 3-Monooxygenase/metabolism
3.
eNeuro ; 9(4)2022.
Article En | MEDLINE | ID: mdl-35701167

The medial forebrain bundle (MFB) is a white matter pathway that traverses through mesolimbic structures and includes dopaminergic neural fibers ascending from the ventral tegmental area (VTA). Since dopaminergic signals represent hedonic responses, electrical stimulation of the MFB in animals has been used as a neural reward for operant and spatial tasks. MFB stimulation strongly motivates animals to rapidly learn to perform a variety of behavioral tasks to obtain a reward. Although the MFB is known to connect various brain regions and MFB stimulation dynamically modulates animal behavior, how central and peripheral functions are affected by MFB stimulation per se is poorly understood. To address this question, we simultaneously recorded electrocorticograms (ECoGs) in the primary motor cortex (M1), primary somatosensory cortex (S1), and olfactory bulb (OB) of behaving rats while electrically stimulating the MFB. We found that MFB stimulation increased the locomotor activity of rats. Spectral analysis confirmed that immediately after MFB stimulation, sniffing activity was facilitated and the power of gamma oscillations in the M1 was increased. After sniffing activity and motor cortical gamma oscillations were facilitated, animals started to move. These results provide insight into the importance of sniffing activity and cortical gamma oscillations for motor execution and learning facilitated by MFB stimulation.


Medial Forebrain Bundle , Motor Cortex , Animals , Dopamine/metabolism , Electric Stimulation , Locomotion , Medial Forebrain Bundle/metabolism , Motor Cortex/metabolism , Rats , Reward , Ventral Tegmental Area/metabolism
4.
Exp Neurol ; 355: 114122, 2022 09.
Article En | MEDLINE | ID: mdl-35636499

BACKGROUND: Deep Brain Stimulation (DBS) of the Medial Forebrain Bundle (MFB) induces antidepressant effects both clinically and pre-clinically. However, the acute electrophysiological changes induced by MFB DBS remain unknown. OBJECTIVE: The study investigated acute mfb DBS effects on neuronal oscillations in distinct neuronal populations implicated in the pathophysiology of depression. METHODS: The Flinders Sensitive Line (FSL) rodent depression model and Sprague-Dawley (SD) controls were used in the study. Recording electrodes were implanted unilaterally in the medial prefrontal cortex (mPFC), nucleus accumbens (NAc), ventral tegmental area (VTA); DBS electrodes were implanted bilaterally in the mfb. The FSL Stim and SD Stim received bilateral mfb DBS, whereas the FSL Sham and SD Shams were not stimulated. Local field potentials (LFPs) from all areas were recorded at baseline, during, and post stimulation. Neuronal oscillations were analyzed. RESULTS: mfb DBS induced 1) a significant increase of low gamma (30-45 Hz) oscillations in the mPFC uniquely in FSLs; 2) a significant increase of low gamma oscillations in the NAc and VTA in SDs and FSLs; and 3) an increase in the expression of Gad1 in the mPFC of FSL and SDs, while only increasing the expression in the NAc of FSLs. CONCLUSION: mfb DBS differentially affected neuronal oscillations in the mPFC, NAc and VTA across SD and FSL rats. Low gamma oscillations rose significantly in the mPFC of FSL rats. Molecular analysis points to a mechanism involving GABAergic interneurons as they regulate low gamma oscillations.


Deep Brain Stimulation , Medial Forebrain Bundle , Animals , Depression/therapy , Medial Forebrain Bundle/metabolism , Rats , Rats, Sprague-Dawley , Rodentia
5.
J Chem Neuroanat ; 113: 101921, 2021 04.
Article En | MEDLINE | ID: mdl-33600923

Human Wharton's jelly-derived Mesenchymal Stromal Cells (hWJ-MSCs) have shown beneficial effects in improving the dopaminergic cells in the Parkinson's disease (PD). In the present study, the effects of hWJ-MSCs on hyperalgesia, anxiety deficiency and Pallidal local electroencephalogram (EEG) impairment, alone and combined with L-dopa, were examined in a rat model of PD. Adult male Wistar rats were divided into five groups: 1) sham, 2) PD, 3) PD + C (Cell therapy), 4) PD + C+D (Drug), and 5) PD + D. PD was induced by injection of 6-OHDA (16 µg/2 µl into medial forebrain bundle (MFB)). PD + C group received hWJ-MSCs (1 × 106 cells, intravenous (i.v.)) twice post PD induction. PD + C+D groups received hWJ-MSCs combined with L-Dopa/Carbidopa, (10/30 mg/kg, intraperitoneally (i.p.)). PD + D group received L-Dopa/Carbidopa alone. Four months later, analgesia, anxiety-like behaviors, were evaluated and Pallidal local EEG was recorded. Level of insulin-like growth factor 1 (IGF-1) was measured in the striatum and dopaminergic neurons were counted in substantia nigra (SNc). According to data, MFB-lesioned rats showed hyperalgesia in tail flick, anxiety-like symptoms in cognitive tests, impairment of electrical power of pallidal local EEG as field potential, count of dopaminergic neurons in SNc and level of IGF-1 in striatum. These complications restored significantly by MSCs treatment (p < 0.001). Our findings confirm that chronic treatment with hWJ-MSC, alone and in combination with L-Dopa, improved nociception and cognitive deficit in PD rats which may be the result of increasing IGF-1 and protect the viability of dopaminergic neurons.


Behavior, Animal/physiology , Mesenchymal Stem Cell Transplantation/methods , Nerve Growth Factor/metabolism , Parkinson Disease, Secondary/therapy , Substantia Nigra/metabolism , Wharton Jelly/cytology , Animals , Antiparkinson Agents/therapeutic use , Carbidopa/therapeutic use , Dopaminergic Neurons/metabolism , Drug Combinations , Electroencephalography , Insulin-Like Growth Factor I/metabolism , Levodopa/therapeutic use , Male , Medial Forebrain Bundle/metabolism , Motor Activity/physiology , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/drug therapy , Parkinson Disease, Secondary/metabolism , Rats , Rats, Wistar
6.
J Neurosci ; 41(2): 234-250, 2021 01 13.
Article En | MEDLINE | ID: mdl-33234607

Dopamine transporter (DAT) controls dopamine neurotransmission by clearing synaptically released dopamine. However, trafficking itineraries of DAT, which determine its cell-surface concentration near synapses, are poorly characterized. It is especially unknown how DAT is transported between spatially distant midbrain somatodendritic and striatal axonal compartments. To examine this "long-range" trafficking, the localization and membrane diffusion of HA-epitope tagged DAT in the medial forebrain bundle (MFB) of a knock-in mouse (both sexes) were analyzed using confocal, super-resolution and EM in intact brain and acute brain slices. HA-DAT was abundant in the plasma membrane of MFB axons, similar to the striatum, although the intracellular fraction of HA-DAT in MFB was more substantial. Intracellular HA-DAT colocalized with VPS35, a subunit of the retromer complex mediating recycling from endosomes, in a subset of axons. Late endosomes, lysosomes, and endoplasmic reticulum were abundant in the soma but minimally present in MFB axons, suggesting that biosynthesis and lysosomal degradation of DAT are confined to soma. Together, the data suggest that membrane diffusion is the main mode of long-range DAT transport through MFB, although the contribution of vesicular traffic can be significant in a population of MFB axons. Based on HA-DAT diffusion rates, plasma membrane DAT in MFB axons turns over with a halftime of ∼20 d, which explains the extremely slow turnover of DAT protein in the brain. Unexpectedly, the mean diameter of DAT-labeled MFB axons was observed to be twice larger than reported for striatum. The implications of this finding for dopamine neuron physiology are discussed.SIGNIFICANCE STATEMENT The dopamine transporter (DAT) is a key regulator of dopamine neurotransmission and a target of abused psychostimulants. In the present study, we examined, for the first time, mechanisms of the long-range traffic of DAT in intact brain and acute brain slices from the knock-in mouse expressing epitope-tagged DAT. Using a combination of confocal, super-resolution and EM, we defined DAT localization and its membrane diffusion parameters in medial forebrain bundle axonal tracts connecting midbrain somatodendritic and striatal axonal compartments of dopaminergic neurons. In contrast to the widely accepted model of long-range axonal transport, our studies suggest that DAT traffics between midbrain and striatum, mainly by lateral diffusion in the plasma membrane with only a limited contribution of vesicular transport in recycling endosomes.


Axons/metabolism , Cell Membrane/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Medial Forebrain Bundle/metabolism , Synaptic Vesicles/metabolism , Animals , Axons/ultrastructure , Diffusion , Dopamine Plasma Membrane Transport Proteins/genetics , Endosomes/metabolism , Female , Gene Knock-In Techniques , Humans , Kinetics , Lysosomes/metabolism , Male , Medial Forebrain Bundle/ultrastructure , Mice , Mice, Inbred C57BL , Vesicular Monoamine Transport Proteins/metabolism
7.
Pharmacol Biochem Behav ; 197: 173013, 2020 10.
Article En | MEDLINE | ID: mdl-32758524

Although extrastriatal dopaminergic (DAergic) systems are being recognized as contributors to Parkinson's disease (PD) pathophysiology, the role of extrastriatal DA depletion in L-Dopa-induced dyskinesia (LID) is still unknown. In view of the physiologic actions of DA on pallidal neuronal activity and the effects on motor behavior of local injection of DA drugs, the loss of the external (GPe, GP in rodents) and internal (GPi, entopeduncular nucleus (EP) in rodents) pallidal DAergic innervation might differentially contribute to LID. A role of pallidal serotonergic (SER) terminals in LID has been highlighted, however, the effect of DAergic innervation is unknown. We investigated the role of DAergic pallidal depletion on LID. Rats were distributed in groups which were concomitantly lesioned with 6-OHDA or vehicle (sham) in the GP, or EP, and in the medial forebrain bundle (MFB) as follows: a) MFB-sham+GP-sham, b) MFB-sham+GP-lesion, c) MFB-lesion+GP-sham, d) MFB-lesion+GP-lesion, e) MFB-sham+EP-sham, f) MFB-sham+EP-lesion, g) MFB-lesion+EP-sham, and h) MFB-lesion+EP-lesion. Four weeks later, animals were treated with L-Dopa (6 mg/kg) twice daily for 22 days.. Immunohistochemical studies were performed in order to investigate the changes in pallidal SER and serotonin transporter (SERT) levels. GP, but not EP, DAergic denervation attenuated LID in rats with a concomitant MFB lesion (p < 0.01). No differences were found in GP SERT expression between groups of animals developing or not LID. These results provide evidence of the relevance of GP DAergic innervation in LID. The conversion of levodopa to DA in GP serotonergic nerve fibers appears not to be the major mechanism underlying LID.


Dyskinesia, Drug-Induced/etiology , Entopeduncular Nucleus/metabolism , Globus Pallidus/metabolism , Levodopa/adverse effects , Oxidopamine/pharmacology , Parkinson Disease, Secondary/chemically induced , Animals , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Entopeduncular Nucleus/drug effects , Entopeduncular Nucleus/physiopathology , Globus Pallidus/drug effects , Globus Pallidus/physiopathology , Levodopa/administration & dosage , Male , Medial Forebrain Bundle/drug effects , Medial Forebrain Bundle/metabolism , Medial Forebrain Bundle/physiopathology , Oxidopamine/administration & dosage , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Signal Transduction/drug effects
8.
Physiol Res ; 69(4): 711-720, 2020 08 31.
Article En | MEDLINE | ID: mdl-32584140

Hypersensitive pain response is observed in patients with Parkinson's disease (PD). However, the signal pathways leading to hyperalgesia still need to be clarified. Chronic oxidative stress is one of the hallmarks of PD pathophysiology. Since the midbrain periaqueductal gray (PAG) is an important component of the descending inhibitory pathway controlling on central pain transmission, we examined the role NADPH oxidase (NOX) of the PAG in regulating exaggerated pain evoked by PD. PD was induced by central microinjection of 6-hydroxydopamine to lesion the left medial forebrain bundle of rats. Then, Western Blot analysis and ELISA were used to determine NOXs and products of oxidative stress (i.e., 8-isoprostaglandin F2alpha and 8-hydroxy-2'-deoxyguanosine). Pain responses to mechanical and thermal stimulation were further examined in control rats and PD rats. In results, among the NOXs, protein expression of NOX4 in the PAG of PD rats was significantly upregulated, thereby the products of oxidative stress were increased. Blocking NOX4 pathway in the PAG attenuated mechanical and thermal pain responses in PD rats and this was accompanied with decreasing production of oxidative stress. In addition, inhibition of NOX4 largely restored the impaired GABA within the PAG. Stimulation of GABA receptors in the PAG of PD rats also blunted pain responses. In conclusions, NOX4 activation of oxidative stress in the PAG of PD rats is likely to impair the descending inhibitory GABAergic pathways in regulating pain transmission and thereby plays a role in the development of pain hypersensitivity in PD. Inhibition of NOX4 has beneficial effects on the exaggerated pain evoked by PD.


Medial Forebrain Bundle/drug effects , NADPH Oxidase 4/antagonists & inhibitors , Pain Threshold/drug effects , Pain/drug therapy , Parkinson Disease/metabolism , Parkinson Disease/pathology , Periaqueductal Gray/drug effects , Pyrazolones/pharmacology , Pyridones/pharmacology , gamma-Aminobutyric Acid/metabolism , Animals , Disease Models, Animal , Male , Medial Forebrain Bundle/metabolism , Pain/etiology , Pain/metabolism , Pain/pathology , Pain Threshold/physiology , Parkinson Disease/enzymology , Periaqueductal Gray/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
9.
Behav Brain Res ; 393: 112786, 2020 09 01.
Article En | MEDLINE | ID: mdl-32592738

Parkinson's disease has become one of the most common neurodegenerative diseases. Pathological changes typically manifest following dopaminergic neuron loss in the substantia nigra and abnormal alpha-synuclein (α-syn) aggregation in the neurons. α-Syn is the major component of Lewy bodies. However, research pertaining to the spread of abnormal α-syn aggregations, which results in specific damage to the brain structure and function, is lacking. In the present study, full-length human α-syn fibrils were injected into the medial forebrain bundle of rats, with an experimental endpoint of 6 months. Histological analysis was conducted to observe the pathological progress of abnormal endogenous α-syn aggregation and nerve fiber quality. Changes in gray and white matter integrity were quantitatively analyzed using voxel-based morphometry (VBM). Behavioral changes were observed over the 6-month period. Histological analysis showed reduced dopamine transporter levels in the striatum of the experimental rats; widespread abnormal endogenous α-syn accumulation; and damaged, sparse, and disordered nerve fibers in the experimental group. VBM showed that at 6 months after surgery, bilateral anterior limbic, bilateral inferior limbic, right hippocampal, and right cortical volumes had reduced, whereas thalamic volume had increased in the experimental group compared with that in the control group. Damage to the limbic and thalamic fiber structure may occur in the earlier stages of Parkinson's disease.


Brain/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Aggregation, Pathological , Serine/metabolism , alpha-Synuclein/metabolism , Animals , Disease Models, Animal , Male , Medial Forebrain Bundle/metabolism , Phosphorylation , Rats, Sprague-Dawley
10.
Exp Neurol ; 327: 113224, 2020 05.
Article En | MEDLINE | ID: mdl-32035070

BACKGROUND: Medial forebrain bundle (MFB) deep brain stimulation (DBS) has anti-depressant effects clinically and in depression models. Currently, therapeutic mechanisms of MFB DBS or how stimulation parameters acutely impact neurotransmitter release, particularly dopamine, are unknown. Experimentally, MFB DBS has been shown to evoke dopamine response in healthy controls, but not yet in a rodent model of depression. OBJECTIVE: The study investigated the impact of clinically used stimulation parameters on the dopamine induced response in a validated rodent depression model and in healthy controls. METHOD: The stimulation-induced dopamine response in Flinders Sensitive Line (FSL, n = 6) rat model of depression was compared with Sprague Dawley (SD, n = 6) rats following MFB DSB, using Fast Scan Cyclic Voltammetry to assess the induced response in the nucleus accumbens. Stimulation parameters were 130 Hz ("clinically" relevant) with pulse widths between 100 and 350 µs. RESULTS: Linear mixed model analysis showed significant impact in both models following MFB DBS both at 130 and 60 Hz with 100 µs pulse width in inducing dopamine response. Furthermore, at 130 Hz the evoked dopamine responses were different across the groups at the different pulse widths. CONCLUSION: The differential impact of MFB DBS on the induced dopamine response, including different response patterns at given pulse widths, is suggestive of physiological and anatomical divergence in the MFB in the pathological and healthy state. Studying how varying stimulation parameters affect the physiological outcome will promote a better understanding of the biological substrate of the disease and the possible anti-depressant mechanisms at play in clinical MFB DBS.


Deep Brain Stimulation/methods , Depression/metabolism , Dopamine/metabolism , Medial Forebrain Bundle/metabolism , Nucleus Accumbens/metabolism , Animals , Depression/physiopathology , Disease Models, Animal , Female , Medial Forebrain Bundle/physiopathology , Nucleus Accumbens/physiopathology , Rats , Rats, Sprague-Dawley
11.
Stereotact Funct Neurosurg ; 98(1): 8-20, 2020.
Article En | MEDLINE | ID: mdl-31982883

BACKGROUND: Deep brain stimulation (DBS) of the medial forebrain bundle (MFB) can reverse depressive-like symptoms clinically and in experimental models of depression, but the mechanisms of action are unknown. OBJECTIVES: This study investigated the role of dopaminergic mechanisms in MFB stimulation-mediated behavior changes, in conjunction with raclopride administration and micropositron emission tomography (micro-PET). METHODS: Flinders Sensitive Line (FSL) rats were allocated into 4 groups: FSL (no treatment), FSL+ (DBS), FSL.R (FSL with raclopride), and FSL.R+ (FSL with raclopride and DBS). Animals were implanted with bilateral electrodes targeting the MFB and given 11 days access to raclopride in the drinking water with or without concurrent continuous bilateral DBS over the last 10 days. Behavioral testing was conducted after stimulation. A PET scan using [18F]desmethoxyfallypride was performed to determine D2 receptor availability before and after raclopride treatment. Changes in gene expression in the nucleus accumbens and the hippocampus were assessed using quantitative polymerase chain reaction. RESULTS: Micro-PET imaging showed that raclopride administration blocked 36% of the D2 receptor in the striatum, but the relative level of blockade was reduced/modulated by stimulation. Raclopride treatment enhanced depressive-like symptoms in several tasks, and the MFB DBS partially reversed the depressive-like phenotype. The raclopride-treated MFB DBS animals had increased levels of mRNA coding for dopamine receptor D1 and D2 suggestive of a stimulation-mediated increase in dopamine receptors. CONCLUSION: Data suggest that chronic and continuous MFB DBS could act via the modulation of the midbrain dopaminergic transmission, including impacting on the postsynaptic dopamine receptor profile.


Deep Brain Stimulation/methods , Depression/metabolism , Dopamine/metabolism , Medial Forebrain Bundle/metabolism , Positron-Emission Tomography/methods , Raclopride/metabolism , Animals , Depression/diagnostic imaging , Depression/therapy , Dopamine Antagonists/metabolism , Dopamine Antagonists/pharmacology , Dopamine Antagonists/therapeutic use , Male , Medial Forebrain Bundle/diagnostic imaging , Medial Forebrain Bundle/drug effects , Raclopride/pharmacology , Raclopride/therapeutic use , Rats , Rodentia/metabolism , X-Ray Microtomography/methods
12.
Brain Res ; 1728: 146595, 2020 02 01.
Article En | MEDLINE | ID: mdl-31830460

Reward deficit, expressed as anhedonia, is one of the major symptoms associated with neuropsychiatric disorders, but the underlying maladaptations have not been understood. Herein, we test the hypothesis that the neuropeptide cocaine- and amphetamine-regulated transcript (CART) may participate in the process. The study is justified since the peptide is a major player in inducing satiety and also processing of reward. The rats were socially isolated to induce reward deficit and conditioned to self-stimulate via an electrode in lateral hypothalamus (LH)-medial forebrain bundle (MFB) region. Compared to group-housed control rats, the socially isolated animals showed decreased lever press activity and elevated ICSS threshold indicating anhedonia-like condition. However, the effects of social isolation were alleviated by CART administered via intracerebroventricular route. The changes in the expression of CART protein and mRNA were screened using immunofluorescence and qRT-PCR methods, respectively. Socially isolated rats showed reduction in the expression of CART in the LH, nucleus accumbens shell (AcbSh) and posterior ventral tegmental area (pVTA) and CART mRNA in the Acb and LH. Double immunostaining with antibodies against CART and synaptophysin revealed significant loss of colabeled elements in LH, AcbSh and pVTA. We suggest that down-regulation of endogenous CARTergic system in the LH-pVTA-AcbSh reward circuitry may be causal to motivational anhedonia like phenotype seen in neuropsychiatric conditions.


Nerve Tissue Proteins/physiology , Reward , Social Isolation , Anhedonia , Animals , Hypothalamic Area, Lateral/metabolism , Locomotion , Male , Medial Forebrain Bundle/metabolism , Motivation , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/genetics , Neuropeptides/metabolism , Rats , Rats, Wistar , Self Stimulation/physiology
13.
Biomolecules ; 9(9)2019 09 01.
Article En | MEDLINE | ID: mdl-31480516

Extensive damage to nigrostriatal dopaminergic neurons leads to Parkinson's disease (PD). To date, the most effective treatment has been administration of levodopa (L-DOPA) to increase dopaminergic tone. This treatment leads to responses that vary widely among patients, from predominantly beneficial effects to the induction of disabling, abnormal movements (L-DOPA induced dyskinesia (LID)). Similarly, experimental studies have shown animals with widely different degrees of LID severity. In this study, unilateral injections of 6-hydroxydopamine (6-OHDA) in the medial forebrain bundle (MFB) produced more than 90% depletion of dopamine in both the striatum and the substantia nigra reticulata (SNr) of rats. Population analysis showed that dopamine depletion levels were clustered in a single population. In contrast, analysis of abnormal involuntary movements (AIMs) induced by L-DOPA treatment of 6-OHDA-lesioned animals yielded two populations: one with mild LID, and the other with severe LID, which are also related to different therapeutic responses. We examined whether the severity of LID correlated with changes in dopamine 3 receptor (D3R) signaling because of the following: (a) D3R expression and the induction of LID are strongly correlated; and (b) dopaminergic denervation induces a qualitative change in D3R signaling in the SNr. We found that the effects of D3R activation on cAMP accumulation and depolarization-induced [3H]-gamma-aminobutyric acid ([3H]-GABA) release were switched. L-DOPA treatment normalized the denervation-induced changes in animals with mild LID. The D3R activation caused depression of both dopamine 1 receptor (D1R)-induced increases in cAMP production and depolarization-induced [3H]-GABA release, which were reversed to their pre-denervation state. In animals with severe LID, none of the denervation-induced changes were reversed. The finding that in the absence of identifiable differences in 6-OHDA and L-DOPA treatment, two populations of animals with different D3R signaling and LIDs severity implies that mechanisms intrinsic to the treated subject determine the segregation.


Dyskinesias/etiology , Dyskinesias/metabolism , Levodopa/adverse effects , Levodopa/therapeutic use , Parkinson Disease/drug therapy , Animals , Cyclic AMP/metabolism , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Male , Medial Forebrain Bundle/drug effects , Medial Forebrain Bundle/metabolism , Oxidopamine/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects
14.
Metab Brain Dis ; 34(6): 1557-1564, 2019 12.
Article En | MEDLINE | ID: mdl-31332728

Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta. The conventional therapeutic measures which include the widely used L-DOPA therapy, are inefficient especially when dopamine loss is severe, and the physical symptoms are full blown. Since neuroinflammation is a core feature of PD, this raised the question of whether early treatment with an anti-inflammatory agent may provide a more efficient intervention for PD. In this study, we investigated the effect of bromelain (an anti-inflammatory drug) on motor responses and dopamine levels in a parkinsonian rat model. Male Sprague-Dawley rats were lesioned stereotaxically with the neurotoxin 6-OHDA. The anti-inflammatory agent, bromelain (40 mg/kg i.p) was used to treat a subset of the rats prior to or 24 h post 6-OHDA lesion. Locomotor activity was assessed after 6-OHDA injection, using the cylinder and step tests. The cortical and striatal concentrations of dopamine were also measured. 6-OHDA injection resulted in marked motor impairment which was prevented by pretreatment with bromelain prior to the lesion. Also, the injection of 6-OHDA into the medial forebrain bundle resulted in a significant reduction in dopamine concentration in the striatum and PFC. Bromelain treatment did not alter the suppression of cortical and striatal dopamine levels. Pre-treatment with bromelain reduced the motor dysfunction in the parkinsonian rat model of PD. The efficacy of treatment with bromelain does not appear to be via preservation of the dopaminergic system. The efficacy of bromelain in 6-OHDA injected rats still remains unclear.


Anti-Inflammatory Agents/therapeutic use , Bromelains/therapeutic use , Medial Forebrain Bundle/drug effects , Motor Activity/drug effects , Parkinson Disease, Secondary/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Behavior, Animal/drug effects , Bromelains/pharmacology , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Male , Medial Forebrain Bundle/metabolism , Medial Forebrain Bundle/physiopathology , Oxidopamine , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/physiopathology , Rats , Rats, Sprague-Dawley
15.
Acta Histochem ; 121(2): 171-181, 2019 Feb.
Article En | MEDLINE | ID: mdl-30573341

Lead (Pb) is a metal element released into the atmosphere and a major source of environmental contamination. The accumulation and concentration of this metal in a food web may lead to the intoxication of the body, more precisely, the nervous system (NS). In addition, Pb-exposure can cause structural and functional disruption of the NS. Studies have shown that Pb-exposure could be a risk factor in the development of Parkinson's disease (PD). The latter is related to dopaminergic deficiency that may be triggered by genetic and environmental factors such as Pb intoxication. In this study, we have evaluated, in one hand, the neurotoxic effect of Pb (25 mg / kg B.W i.p) for three consecutive days on dopaminergic system and locomotor performance in Merione shawi. In the other hand, the possible restorative potential of C. sativus (CS) (50 mg / kg BW) by oral gavage. The immunohistochemical approach has revealed that Pb-intoxicated Meriones show a significant increase of Tyrosine Hydroxylase (TH) levels within the Substantia Nigra compacta (SNc), Ventral Tegmental Area (VTA), Locus Coeruleus (LC), Dorsal Striatum (DS) and Medial Forebrain Bundle (MFB), unlike the control meriones, a group intoxicated and treated with Crocus sativus hydroethanolic extract (CSHEE) and treated group by CSHEE. Treatment with CSHEE, has shown a real potential to prevent all Pb-induced damages. In fact, restores the TH levels by 92%, 90%, 88%, 90% and 93% in SNc, VTA, LC, DS and MFB respectively, similarly, locomotor activity dysfunction in Pb-intoxicaed meriones was reinstated by 90%. In this study, we have revealed a new pharmacological potential of Crocus sativus that can be used as a neuroprotective product for neurodegenerative disorders, especially, which implying dopaminergic and noradrenergic injuries, like PD, trigged by heavy metals.


Dopaminergic Neurons/metabolism , Parkinson Disease/metabolism , Pars Compacta/metabolism , Ventral Tegmental Area/metabolism , Animals , Crocus/metabolism , Dopamine/metabolism , Male , Medial Forebrain Bundle/metabolism , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism
16.
Free Radic Biol Med ; 129: 73-87, 2018 12.
Article En | MEDLINE | ID: mdl-30176346

Wnts and the components of Wnt/ß-catenin signaling are widely expressed in midbrain and required to control the fate specification of dopaminergic (DAergic) neurons, a neuronal population that specifically degenerate in Parkinson's disease (PD). Accumulating evidence suggest that mitochondrial dysfunction plays a key role in pathogenesis of PD. Axin-2, a negative regulator of Wnt/ß-catenin signaling affects mitochondrial biogenesis and death/birth of new DAergic neurons is not fully explored. We investigated the functional role of Axin-2/Wnt/ß-catenin signaling in mitochondrial biogenesis and DAergic neurogenesis in 6-hydroxydopamine (6-OHDA) induced rat model of PD-like phenotypes. We demonstrate that single unilateral injection of 6-OHDA into the medial forebrain bundle (MFB) potentially dysregulates Wnt/ß-catenin signaling in substantia nigra pars compacta (SNpc). We used shRNA lentiviruses to genetically knockdown Axin-2 to up-regulate Wnt/ß-catenin signaling in SNpc in parkinsonian rats. Genetic knockdown of Axin-2 up-regulates Wnt/ß-catenin signaling by destabilizing the ß-catenin degradation complex in SNpc in parkinsonian rats. Axin-2 shRNA mediated activation of Wnt/ß-catenin signaling improved behavioural functions and protected the nigral DAergic neurons by increasing mitochondrial functionality in parkinsonian rats. Axin-2 shRNA treatment reduced apoptotic signaling, autophagy and ROS generation and improved mitochondrial membrane potential which promotes mitochondrial biogenesis in SNpc in parkinsonian rats. Interestingly, Axin-2 shRNA-mediated up-regulation of Wnt/ß-catenin signaling enhanced net DAergic neurogenesis by regulating proneural genes (Nurr-1, Pitx-3, Ngn-2, and NeuroD1) and mitochondrial biogenesis in SNpc in parkinsonian rats. Therefore, our data suggest that pharmacological/genetic manipulation of Wnt signaling that enhances the endogenous regenerative capacity of DAergic neurons may have implication for regenerative approaches in PD.


Adaptor Proteins, Signal Transducing/genetics , Carrier Proteins/genetics , Dopaminergic Neurons/metabolism , Neurogenesis/genetics , Parkinson Disease, Secondary/genetics , Wnt Proteins/genetics , Wnt Signaling Pathway , beta Catenin/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Injections, Intraventricular , Male , Medial Forebrain Bundle/drug effects , Medial Forebrain Bundle/metabolism , Medial Forebrain Bundle/pathology , Mesencephalon/drug effects , Mesencephalon/metabolism , Mesencephalon/pathology , Mitochondria/genetics , Mitochondria/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Organelle Biogenesis , Oxidopamine/administration & dosage , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Pars Compacta/drug effects , Pars Compacta/metabolism , Pars Compacta/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Stereotaxic Techniques , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
17.
Brain Res ; 1673: 52-63, 2017 Oct 15.
Article En | MEDLINE | ID: mdl-28760440

Electroacupuncture (EA) has been reported to alleviate motor deficits in Parkinson's disease (PD) patients, and PD animal models. However, the mechanisms by which EA improves motor function have not been investigated. We have employed a 6-hydroxydopamine (6-OHDA) unilateral injection induced PD model to investigate whether EA alters protein expression in the motor cortex. We found that 4weeks of EA treatment significantly improved spontaneous floor plane locomotion and rotarod performance. High-throughput proteomic analysis in the motor cortex was employed. The expression of 54 proteins were altered in the unlesioned motor cortex, and 102 protein expressions were altered in the lesioned motor cortex of 6-OHDA rats compared to sham rats. Compared to non-treatment PD control, EA treatment reversed 6 proteins in unlesioned and 19 proteins in lesioned motor cortex. The present study demonstrated that PD induces proteomic changes in the motor cortex, some of which are rescued by EA treatment. These targeted proteins were mainly involved in increasing autophagy, mRNA processing and ATP binding and maintaining the balance of neurotransmitters.


Electroacupuncture , Motor Cortex/metabolism , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/therapy , Proteome , Animals , Apomorphine/pharmacology , Blotting, Western , Chromatography, High Pressure Liquid , Dopamine Agonists/pharmacology , Immunohistochemistry , Male , Mass Spectrometry , Medial Forebrain Bundle/metabolism , Medial Forebrain Bundle/pathology , Motor Activity/drug effects , Motor Activity/physiology , Oxidopamine , Parkinsonian Disorders/pathology , Proteomics , Random Allocation , Rats, Sprague-Dawley , Tyrosine 3-Monooxygenase/metabolism
18.
J Affect Disord ; 217: 80-88, 2017 08 01.
Article En | MEDLINE | ID: mdl-28395208

BACKGROUND: Among several potential neuroanatomical targets pursued for deep brain stimulation (DBS) for treating those with treatment-resistant depression (TRD), the superolateral-branch of the medial forebrain bundle (MFB) is emerging as a privileged location. We investigated the antidepressant-like phenotypic and chemical changes associated with reward-processing dopaminergic systems in rat brains after MFB-DBS. METHODS: Male Wistar rats were divided into three groups: sham-operated, DBS-Off, and DBS-On. For DBS, a concentric bipolar electrode was stereotactically implanted into the right MFB. Exploratory activity and depression-like behavior were evaluated using the open-field and forced-swimming test (FST), respectively. MFB-DBS effects on the dopaminergic system were evaluated using immunoblotting for tyrosine hydroxylase (TH), dopamine transporter (DAT), and dopamine receptors (D1-D5), and high-performance liquid chromatography for quantifying dopamine, 3,4-dihydroxyphenylacetic acid (DOPAC), and homovanillic acid (HVA) in brain homogenates of prefrontal cortex (PFC), hippocampus, amygdala, and nucleus accumbens (NAc). RESULTS: Animals receiving MFB-DBS showed a significant increase in swimming time without alterations in locomotor activity, relative to the DBS-Off (p<0.039) and sham-operated groups (p<0.014), indicating an antidepressant-like response. MFB-DBS led to a striking increase in protein levels of dopamine D2 receptors and DAT in the PFC and hippocampus, respectively. However, we did not observe appreciable differences in the expression of other dopamine receptors, TH, or in the concentrations of dopamine, DOPAC, and HVA in PFC, hippocampus, amygdala, and NAc. LIMITATIONS: This study was not performed on an animal model of TRD. CONCLUSION: MFB-DBS rescues the depression-like phenotypes and selectively activates expression of dopamine receptors in brain regions distant from the target area of stimulation.


Depressive Disorder, Treatment-Resistant/therapy , Dopamine Plasma Membrane Transport Proteins/metabolism , Medial Forebrain Bundle/metabolism , Receptors, Dopamine D2/metabolism , Animals , Antidepressive Agents/metabolism , Brain/metabolism , Deep Brain Stimulation/methods , Depressive Disorder, Treatment-Resistant/metabolism , Dopamine/metabolism , Male , Prefrontal Cortex/metabolism , Rats , Rats, Wistar
19.
Metab Brain Dis ; 32(4): 1061-1067, 2017 08.
Article En | MEDLINE | ID: mdl-28321600

Chronic exposure to stress amplifies locomotor deficits and exacerbates dopamine neuron loss in an animal model for Parkinson's disease. The release of neurotrophic factors such as glial cell-line derived neurotrophic factor (GDNF) and neurotrophin-3 (NT-3) following neuronal injury attenuates exacerbated degeneration of these neurons. In this study, the neurotoxin 6-hydroxydopamine (6-OHDA) was injected unilaterally into the medial forebrain bundle of male Sprague Dawley rats. A subset of these rats was subjected to post-lesion restraint stress after which the effect of exposure to stress on locomotor activity (forelimb akinesia test), neurotrophic factor (GDNF and NT-3) and corticosterone concentration was assessed. Exposure to post-lesion stress resulted in increased preference to use the unimpaired forelimb (forelimb ipsilateral to the lesioned hemisphere) in the forelimb akinesia test. The expected increase in both GDNF and NT-3 concentration following injury was not present in the stressed animals. However, both the non-stressed and stressed lesioned groups had decreased neurotrophic factor concentration at one and two weeks post lesion. This decrease was exaggerated in the stressed rats. The decrease in neurotrophic factor concentration was accompanied by an increase in corticosterone concentration in the stressed rats. These findings demonstrate that exposure to post-6-OHDA lesion stress exaggerates dopamine neurodegeneration and enhance motor impairment. This suggests that conditions that result in a hyper-activated hypothalamic-pituitary-adrenal axis such as depression which is concomitant to a Parkinson's disease diagnosis may be responsible for enhanced dopamine depletion by attenuating neurotrophic factor concentration elevation in the nigrostriatal pathway following neuronal injury.


Corpus Striatum/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Medial Forebrain Bundle/metabolism , Motor Skills/physiology , Neurotrophin 3/metabolism , Stress, Physiological/physiology , Stress, Psychological/metabolism , Animals , Corticosterone/blood , Hypothalamo-Hypophyseal System/metabolism , Male , Oxidopamine , Pituitary-Adrenal System/metabolism , Rats , Rats, Sprague-Dawley , Restraint, Physical
20.
Exp Neurol ; 292: 11-20, 2017 06.
Article En | MEDLINE | ID: mdl-28223037

Asymmetric dopamine loss is commonly found in early Parkinson's disease (PD), but its effects on functional networks have been difficult to delineate in PD patients because of variations in age, disease duration and therapy. Here we used unilateral 6-hydroxydopamine-lesioned (6-OHDA) rats and controls and treated them with a single intraperitoneal injection of levodopa (L-DOPA) before performing diffusion weighted MRI and resting state functional MRI (rs-fMRI). In accordance with a neurodegeneration of the nigrostriatal dopaminergic pathway, diffusion tensor imaging showed increased radial diffusivity and decreased fractional anisotropy in the lesioned substantia nigra. Likewise a deterministic connectometry approach showed increase of isotropic diffusion values in the medial forebrain bundle. rs-fMRI showed reduced interhemispheric functional connectivity (FC) between the intact and the 6-OHDA lesioned caudate-putamen. Unexpectedly, there was an increased FC between the 6-OHDA lesioned caudate-putamen and sensorimotor cortices of both hemispheres. L-DOPA reversed the FC changes between the dopamine denervated caudate-putamen and the sensorimotor cortices, but not the reduced interhemispheric FC between caudate-putamina. Similarly, L-DOPA induced c-fos expression in both sensorimotor cortices, but only in the dopamine-depleted caudate-putamen. Taken together, these data suggest that asymmetric degeneration of the nigrostriatal dopamine pathway results in functional asynchrony between the intact and 6-OHDA-lesioned caudate-putamen and increased interhemispheric synchrony between sensorimotor cortices. The results also indicate that the initial effect of L-DOPA is to restore functional corticostriatal connectivity rather than synchronize caudate-putamina.


Corpus Striatum/drug effects , Dopamine/metabolism , Levodopa/pharmacology , Parkinsonian Disorders/physiopathology , Substantia Nigra/drug effects , Animals , Corpus Striatum/metabolism , Diffusion Tensor Imaging/methods , Disease Models, Animal , Male , Medial Forebrain Bundle/drug effects , Medial Forebrain Bundle/metabolism , Parkinsonian Disorders/drug therapy , Rats, Sprague-Dawley , Substantia Nigra/metabolism
...