Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
Neurochem Int ; 148: 105066, 2021 09.
Article in English | MEDLINE | ID: mdl-34004240

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder that affects more than 10 million people worldwide. Oxidative stress and mitochondrial dysfunction play a significant role in altering the homeostasis of energy production and free radical generation. Current PD therapies are focused on reducing the cardinal symptoms rather than preventing disease progression in the patients. Adenosine A2A receptor (A2A R) antagonist (Istradephylline) combined with levodopa shows a promising therapy for PD. In animal studies, caffeine administration showed to improve motor functions and neuroprotective effect in the neurons. Caffeine is probably the most extensively used psychoactive substance. In this current study, we investigated the neuroprotective effect of caffeine against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurodegeneration. Here, we demonstrate that caffeine improves behavioral and neurotransmitter recovery against MPTP-induced toxicity. Caffeine restores endogenous antioxidant levels and suppresses neuroinflammation. Our finding suggests that the blockage of A2AR is a promising disease-modifying therapy for PD. Target engagement strategies could be more beneficial in preventing disease progression rather than symptomatic reliefs in PD patients.


Subject(s)
Caffeine/pharmacology , Dietary Supplements , Dopaminergic Neurons/drug effects , MPTP Poisoning/drug therapy , Neuroprotective Agents/pharmacology , Animals , Antioxidants/pharmacology , Behavior, Animal/drug effects , MPTP Poisoning/pathology , MPTP Poisoning/psychology , Mice , Mice, Inbred C57BL , Neuroinflammatory Diseases/drug therapy , Substantia Nigra/metabolism , Substantia Nigra/pathology
2.
Neuropharmacology ; 182: 108386, 2021 01.
Article in English | MEDLINE | ID: mdl-33152452

ABSTRACT

In Parkinson's disease (PD), management of L-3,4-dihydroxyphenylalanine (l-DOPA)-related complications, such as l-DOPA induced dyskinesia and psychosis, remains inadequate, which poses a significant burden on the quality of life of patients. We have shown, in the hemi-parkinsonian rat model of PD, that the selective serotonin type 3 (5-HT3) receptor antagonists ondansetron and granisetron decreased the severity of established dyskinesia, and ondansetron even attenuated the development of dyskinesia. Here, we seek to confirm these favourable data on dyskinesia and to explore the effect of ondansetron on the severity of psychosis-like behaviours (PLBs) in the gold standard model of PD, the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned non-human primate. We first determined the pharmacokinetic profile of ondansetron in the marmoset. Subsequently, six MPTP-lesioned marmosets were administered l-DOPA chronically until they exhibited stable and reproducible dyskinesia and PLBs upon each administration of l-DOPA. On behavioural assessment days, ondansetron (0.01, 0.1 and 1 mg/kg) or vehicle was administered in conjunction with l-DOPA, and the severity of dyskinesia, PLBs and parkinsonism was evaluated. Ondansetron 0.1 mg/kg alleviated global dyskinesia severity by 73% (P < 0.0001) and decreased duration of on-time with disabling dyskinesia by 88% (P = 0.0491). Ondansetron 0.1 mg/kg reduced the severity of global PLBs by 80% (P < 0.0001) and suppressed on-time with disabling PLBs (P = 0.0213). Ondansetron enhanced the anti-parkinsonian action of l-DOPA, reducing global parkinsonism by 53% compared to l-DOPA (P = 0.0004). These results suggest that selective blockade of the 5-HT3 receptor with ondansetron may be an effective approach to alleviate l-DOPA-related complications.


Subject(s)
Anti-Anxiety Agents/therapeutic use , MPTP Poisoning/drug therapy , Ondansetron/therapeutic use , Psychotic Disorders/drug therapy , Serotonin 5-HT3 Receptor Antagonists/therapeutic use , Animals , Callithrix , Dose-Response Relationship, Drug , Female , MPTP Poisoning/physiopathology , MPTP Poisoning/psychology , Male , Psychotic Disorders/physiopathology , Psychotic Disorders/psychology
3.
Am J Physiol Endocrinol Metab ; 319(4): E734-E743, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32865008

ABSTRACT

Autophagy dysfunctions are involved in the pathogenesis of Parkinson's disease (PD). In the present study, we aimed to evaluate the involvement of G protein-coupled estrogen receptor (GPER) in the inhibitory effect of insulin-like growth factor-1 (IGF-1) against excessive autophagy in PD animal and cellular models. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment significantly induced mouse movement disorder and decreased the protein level of tyrosine hydroxylase (TH) in the substantia nigra (SN) and dopamine (DA) content in striatum. Along with the dopamine neuron injury, we observed significant upregulations of microtubule-associated light chain-3 II (LC3-II) and α-synuclein as well as a downregulation of P62 in MPTP-treated mice. These changes could be restored by IGF-1 pretreatment. Cotreatment with IGF-1R antagonist JB-1 or GPER antagonist G15 could block the neuroprotective effects of IGF-1. 1-Methy-4-phenylpyridinium (MPP+) treatment could also excessively activate autophagy along with the reduction of cell viability in SH-SY5Y cells. IGF-1 could inhibit the neurotoxicity through promoting the phosphorylation of Akt and mammalian target of rapamycin (mTOR), which could also be antagonized by JB-1 or G15. These data suggest that IGF-1 inhibits MPTP/MPP+-induced autophagy on dopaminergic neurons through the IGF-1R/PI3K-Akt-mTOR pathway and GPER.


Subject(s)
Autophagy/drug effects , Dopaminergic Neurons/drug effects , Insulin-Like Growth Factor I/pharmacology , MPTP Poisoning/prevention & control , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects , Animals , Humans , MPTP Poisoning/psychology , Male , Mice , Mice, Inbred C57BL , Neostriatum/drug effects , Neostriatum/metabolism , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Postural Balance/drug effects , Receptor, IGF Type 1 , TOR Serine-Threonine Kinases/metabolism
4.
J Alzheimers Dis ; 74(3): 883-901, 2020.
Article in English | MEDLINE | ID: mdl-32083584

ABSTRACT

Neurotoxicity is one of the major pathological changes in multiple neurological disorders, including Alzheimer's disease (AD) and Parkinson's disease (PD), the second popular neurodegenerative disease in aged people. It is known that the AD and PD share the similar neuropathological hallmarks, such as the oxidative stress, loss of specific neurons, and aggregation of specific proteins. However, there are no effective therapeutic drugs for both AD and PD yet. Oxytocin (OXT) is a small peptide with 9 amino acids that is neuroprotective to many neurological disorders. Whether OXT administration confers neuroprotection to 1-methyl-4-phenyl-1, 2, 3, 6- tetrahydropyridine (MPTP)-induced neurotoxicity in mice are still not known. In this study, we first found that the OXT levels are decreased in MPTP mice. Supplementation with OXT effectively rescues the locomotor disabilities and anxiety-like behaviors in MPTP mice. OXT also alleviates the hyperphosphorylation of α-synuclein at S129 site and the loss of dopaminergic neurons in the substantia nigra pars compacta, as well as the oxidative stress in the MPTP mice, and alleviates both oxidative stress and cell cytotoxicity in vitro. Furthermore, we found that OXT could inhibit the miR-26a/DAPK1 signal pathway in MPTP mice. In summary, our study demonstrates protective effects of OXT in MPTP mice and that miR-26a/DAPK1 signaling pathway may play an important role in mediating the protection of OXT.


Subject(s)
Death-Associated Protein Kinases/drug effects , MPTP Poisoning/drug therapy , MicroRNAs/drug effects , Neuroprotective Agents/therapeutic use , Oxytocin/therapeutic use , Signal Transduction/drug effects , Animals , Behavior, Animal/drug effects , Cell Line, Tumor , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , MPTP Poisoning/psychology , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Oxidative Stress/drug effects , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/psychology , Psychomotor Performance/drug effects
5.
Eur J Pharmacol ; 873: 172957, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32004527

ABSTRACT

Psychosis and dyskinesia significantly diminish the quality of life of patients with advanced Parkinson's disease (PD). Available treatment options are unfortunately few and their use is limited by adverse effects. We have recently shown that activation of metabotropic glutamate 2 and 3 (mGlu2/3) receptors produced significant relief of L-3,4-dihydroxyphenylalanine (L-DOPA)-induced psychosis-like behaviours (PLBs) and dyskinesia in experimental models of PD. Here, using the highly-selective mGlu2 positive allosteric modulator (PAM) LY-487,379, we seek to determine the contribution of selective mGlu2 activation on both L-DOPA-induced PLBs and dyskinesia, in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned primate. We first determined the pharmacokinetic (PK) profile of LY-487,379 in the common marmoset, following which we administered it (0.1, 1 and 10 mg/kg) or its vehicle to 6 MPTP-lesioned marmosets previously exposed to L-DOPA to elicit stable PLBs and dyskinesia. We found that LY-487,379 provided a ≈45% reduction of the global PLBs observed and reduced global dyskinesia score by ≈ 55%. Moreover, LY-487,379 enhanced the anti-parkinsonian effect of L-DOPA, by reducing global parkinsonian score by ≈ 15%. Our data suggest that selective mGlu2 positive allosteric modulation with LY-487,379 may represent a potential therapeutic approach to alleviate both L-DOPA-induced PLBs and dyskinesia in PD.


Subject(s)
Antipsychotic Agents/therapeutic use , Behavior, Animal/drug effects , Dyskinesia, Drug-Induced/drug therapy , GABA Modulators/therapeutic use , Levodopa , MPTP Poisoning/drug therapy , Psychoses, Substance-Induced/drug therapy , Pyridines/therapeutic use , Receptors, Metabotropic Glutamate/drug effects , Sulfonamides/therapeutic use , Animals , Antiparkinson Agents/pharmacology , Antiparkinson Agents/therapeutic use , Callithrix , Female , GABA Modulators/pharmacokinetics , MPTP Poisoning/psychology , Male , Parkinson Disease/complications , Parkinson Disease/drug therapy , Parkinson Disease/psychology , Psychoses, Substance-Induced/psychology , Pyridines/pharmacokinetics , Sulfonamides/pharmacokinetics
6.
Toxicology ; 426: 152256, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31381935

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative disorder that is characterized by the degeneration of dopaminergic neurons in substantia nigra (SN). Accumulating evidences implicate the beneficial role of estrogen in the therapy of PD. METHODS: In the present study, the protective function of luteolin-7-O-glucoside (LUT-7G), a natural flavonoid, was investigated in 1-methyl-4-phenylpyridinium (MPP+) treated SH-SY5Y cells and 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) induced mice. RESULTS: Pre-treatment of LUT-7G increased the viability and reduced the apoptosis of SH-SY5Y cells treated by MPP+. At molecular level, the Bcl-2/Bax ratio was increased, while the expression of cleaved caspase 3 was markedly lessened. Moreover, LUT-7G increased the expression of estrogen receptor (ER), ERα and ERß, and enhanced the activation of ERK1/2/STAT3/c-Fos that could be abolished by ER antagonists. Furthermore, in vivo experiment indicated that pre-treatment of LUT-7G improved the bradykinesia, and enhanced the muscle strength as well as the balancing capacity of mice treated with MPTP. And LUT-7G prevented the injury of TH positive cells in substantia nigra and increased TH positive nerve fibers in striatum. In addition, pre-treatment of LUT-7G also significantly diminished the MPTP-induced gliosis in substantia nigra. CONCLUSIONS: LUT-7G effectively protected dopaminergic neurons against MPP+ or MPTP-induced toxicity, probably by activating the ER-mediated signaling pathway. Our findings explore the therapeutic potential of LUT-7G for PD therapy.


Subject(s)
Dopaminergic Neurons/drug effects , Flavones/pharmacology , Glucosides/pharmacology , MPTP Poisoning/prevention & control , Neuroprotective Agents/pharmacology , Receptors, Estrogen/drug effects , Signal Transduction/drug effects , Animals , Behavior, Animal/drug effects , Cell Line , Estrogen Antagonists/pharmacology , Humans , Hypokinesia/etiology , Hypokinesia/prevention & control , MAP Kinase Signaling System/drug effects , MPTP Poisoning/pathology , MPTP Poisoning/psychology , Male , Mice , Mice, Inbred C57BL , Muscle Strength/drug effects , STAT3 Transcription Factor/drug effects , Substantia Nigra/pathology
7.
Aging (Albany NY) ; 11(7): 1934-1964, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30958793

ABSTRACT

Memory deficiency is a common non-motor symptom of Parkinson's disease (PD), and conventionally, α-synuclein is considered to be an important biomarker for both motor and cognitive characteristics attributed to PD. However, the role of physiological α-synuclein in cognitive impairment remains undetermined. Ginsenoside Rb1 has been shown to protect dopaminergic neurons (DA) from death and inhibit α-synuclein fibrillation and toxicity in vitro. Our recent study also revealed that ginsenoside Rb1 ameliorates motor deficits and prevents DA neuron death via upregulating glutamate transporter GLT-1 in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Whether Rb1 can improve memory deficiency and the underlying mechanism is still unknown. In this study, we found that Rb1 can prevent the spatial learning and memory deficits, increase long-term potentiation (LTP) and hippocampal glutamatergic transmission in the MPTP mouse model. The underlying neuroprotective mechanism of Rb1-improved synaptic plasticity involves Rb1 promoting hippocampal CA3 α-synuclein expression, restoring the glutamate in the CA3-schaffer collateral-CA1 pathway, and sequentially increasing postsynaptic density-95 (PSD-95) expression. Thus, we provide evidence that Rb1 modulates memory function, synaptic plasticity, and excitatory transmission via the trans-synaptic α-synuclein/PSD-95 pathway. Our findings suggest that Rb1 may serve as a functional drug in treating the memory deficiency in PD.


Subject(s)
Disks Large Homolog 4 Protein/metabolism , Ginsenosides/pharmacology , Hippocampus/drug effects , Hippocampus/physiology , Memory/drug effects , Memory/physiology , alpha-Synuclein/metabolism , Animals , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/physiology , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques , MPTP Poisoning/drug therapy , MPTP Poisoning/physiopathology , MPTP Poisoning/psychology , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/physiopathology , Parkinsonian Disorders/psychology , Signal Transduction/drug effects , Synaptic Transmission/drug effects , alpha-Synuclein/antagonists & inhibitors , alpha-Synuclein/genetics
8.
Behav Brain Res ; 369: 111915, 2019 09 02.
Article in English | MEDLINE | ID: mdl-30998993

ABSTRACT

The neurodegenerative disorder Parkinson's disease affects motor abilities as well as cognition. The gold standard therapy is L-Dopa, which mainly restores motor skills. Therefore, we require additional interventions to sustain cognitive functions in Parkinson's disease. The lifestyle intervention "physical activity" improves adult hippocampal neurogenesis and memory but so far, its impact has not been investigated in rodent models for Parkinson's disease previously treated with the standard therapy. We hereby asked whether physical activity serves as a pro-neurogenic and -cognitive stimulus in dopamine-depleted mice previously treated with L-Dopa. Therefore, we injected dopamine-depleted mice with L-Dopa/Benserazide followed either by exercise or by a sedentary lifestyle. We analysed adult hippocampal neurogenesis histologically and assessed spatial memory in the Morris water maze. Furthermore, we investigated the hippocampal and striatal monoaminergic cross-talk. Physical activity prevented memory decline and was linked to a slower dopamine turnover but did not enhance neurogenesis in dopamine-depleted mice previously treated with L-Dopa. In conclusion, physical activity did not develop its full pro-neurogenic potential in mice previously treated with L-Dopa but sustained spatial cognition in Parkinson's disease.


Subject(s)
Antiparkinson Agents/pharmacology , Benserazide/pharmacology , Hippocampus/physiopathology , Levodopa/pharmacology , MPTP Poisoning/therapy , Memory/physiology , Motor Activity/physiology , Animals , Antiparkinson Agents/adverse effects , Benserazide/adverse effects , Corpus Striatum/drug effects , Corpus Striatum/physiopathology , Disease Models, Animal , Dopamine/metabolism , Drug Combinations , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Levodopa/adverse effects , MPTP Poisoning/pathology , MPTP Poisoning/physiopathology , MPTP Poisoning/psychology , Maze Learning/drug effects , Maze Learning/physiology , Memory/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Neurogenesis/drug effects , Neurogenesis/physiology , Random Allocation , Sedentary Behavior
9.
Neurotox Res ; 34(4): 808-819, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29808370

ABSTRACT

Depression is a highly prevalent and debilitating non-motor symptom observed during the early stages of Parkinson's disease (PD). Although PD prevalence is higher in men, the depressive symptoms in PD are more common in women. Therefore, the aim of this study was to investigate the development of anhedonic- and depressive-like behaviors in male and female mice and the potential mechanisms related to depressive symptoms in an experimental model of PD. Young adult male and female C57BL/6 mice (3 months old) received a single intranasal (i.n.) administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and were submitted to a battery of behavioral tasks (sucrose consumption, splash test, tail suspension, forced swimming and open field tests) to assess their emotional and motor profiles. Considering the role of sexual hormones in emotional behaviors, the same protocol of i.n. MPTP administration and the splash, tail suspension, and open field tests were conducted in ovariectomized (OVX) and aged C57BL/6 female (20 months old) mice. We also investigated the immunocontent of neurotrophins (BDNF, GDNF, and VEGF) in the hippocampus and prefrontal cortex by western blot. I.n.  MPTP administration induced more pronounced anhedonic- and selective depressive-like behaviors in female adult mice, also observed in OVX and aged female mice, with the absence of motor impairments. Furthermore, MPTP induced a more pronounced depletion of neurotrophins in the prefrontal cortex and hippocampus in female than male mice. This study provides new evidence of increased susceptibility of female mice to anhedonic- and depressive-like behaviors following i.n. MPTP administration. The observed gender-related effects of MPTP on emotional parameters seem to be linked to increased depletion of neurotrophins (particularly BDNF and GDNF) in the hippocampus and prefrontal cortex of female mice.


Subject(s)
Anhedonia/physiology , Depression/physiopathology , MPTP Poisoning/physiopathology , MPTP Poisoning/psychology , Administration, Intranasal , Aging/physiology , Anhedonia/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Depression/chemically induced , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Ovariectomy , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Sex Factors , Tyrosine 3-Monooxygenase/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
J Neural Transm (Vienna) ; 125(9): 1355-1360, 2018 09.
Article in English | MEDLINE | ID: mdl-29247391

ABSTRACT

Trazodone is a clinically available anti-depressant that exhibits affinity for serotonin 1A and 2A receptors, as well as for alpha-adrenoceptors, suggesting that it may be useful to treat L-3,4-dihydroxyphenylalanine (L-DOPA)-induced dyskinesia and psychosis that are encountered in advanced Parkinson's disease (PD). Here, we investigated the anti-dyskinetic and anti-psychotic effects of trazodone in the parkinsonian non-human primate. 6 common marmosets were rendered parkinsonian by administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Following repeated administration of L-DOPA to induce stable dyskinesia and psychosis-like behaviours (PLBs), trazodone (0.1, 1 and 10 mg/kg) or vehicle was administered in combination with L-DOPA and its effects on dyskinesia, PLBs and parkinsonism were determined. The addition of trazodone 10 mg/kg to L-DOPA reduced peak dose dyskinesia by ≈ 39% (P < 0.01) and peak dose PLBs by ≈ 17% (P < 0.01). However, parkinsonian disability was significantly worsened by trazodone 10 mg/kg (P < 0.05) and duration of anti-parkinsonian action was diminished by ≈ 21% (P < 0.05). Our results suggest that trazodone may be effective in alleviating L-DOPA-induced dyskinesia and psychosis in PD, but its deleterious effect on motor function is a concern and may limit its tolerability and usefulness in clinical settings.


Subject(s)
Antiparkinson Agents/toxicity , Dyskinesia, Drug-Induced/drug therapy , Levodopa/toxicity , MPTP Poisoning/physiopathology , Parkinsonian Disorders/physiopathology , Serotonin Receptor Agonists/therapeutic use , Trazodone/therapeutic use , Animals , Antiparkinson Agents/therapeutic use , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Behavior, Animal/drug effects , Callithrix , Disorders of Excessive Somnolence/chemically induced , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/prevention & control , Female , Levodopa/therapeutic use , MPTP Poisoning/drug therapy , MPTP Poisoning/psychology , Male , Motor Activity/drug effects , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/psychology , Serotonin Receptor Agonists/pharmacology , Trazodone/pharmacology , Trazodone/toxicity
11.
Arch Physiol Biochem ; 124(4): 292-299, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29096532

ABSTRACT

BACKGROUND: Parkinson's disease is a progressive neurodegenerative disorder. Aspartame (l-aspartyl-l-phenylalanine methyl ester), a low calorie sweetener used in foods and beverages. OBJECTIVES: This study investigated the effect of chronic aspartame intake on Parkinsonism induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). METHOD: Forty-eight mice (24 males and 24 females): control, aspartame, MPTP, and aspartame + MPTP groups tested by Y-maze, stepping, forced swimming and olfactory preference tests. Brain tissues examined for dopamine content, tyrosine hydroxylase, inducible nitric oxide synthase (iNOS), glutathione peroxidase, phosphorylated tau and α-synuclein protein. Histopathological evaluation of brain sections at the level of basal ganglia was done. RESULTS: Decreased dopamine content, tyrosine hydroxylase expression, glutathione peroxidase expression and increased iNOS, tau and α-synuclein expression in groups received aspartame, MPTP or both agents simultaneously in both males and females group. CONCLUSIONS: Increased dopaminergic degeneration and complications with chronic aspartame consumption and more injury in male groups.


Subject(s)
Aspartame/adverse effects , Basal Ganglia/metabolism , Dopamine Antagonists/adverse effects , Dopaminergic Neurons/metabolism , Gene Expression Regulation , MPTP Poisoning/metabolism , Non-Nutritive Sweeteners/adverse effects , Agnosia/etiology , Animals , Basal Ganglia/enzymology , Basal Ganglia/pathology , Behavior, Animal , Cognitive Dysfunction/etiology , Depression/etiology , Disease Progression , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/pathology , Female , Learning Disabilities/etiology , MPTP Poisoning/pathology , MPTP Poisoning/physiopathology , MPTP Poisoning/psychology , Male , Mice , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Sex Characteristics , Toxicity Tests, Chronic
12.
Biochem Biophys Res Commun ; 493(4): 1443-1449, 2017 12 02.
Article in English | MEDLINE | ID: mdl-28986252

ABSTRACT

Uric acid has neuroprotective effect on Parkinson's disease (PD) by inhibiting oxidative damage and neuronal cell death. Our previous study has shown that uric acid protected dopaminergic cell line damage through inhibiting accumulation of NF-E2-related factor 2 (Nrf2). This study aimed to investigate its in vivo neuroprotective effect. PD was induced by MPTP intraperitoneally injection for 7 d in male C57BL/6 mice. Mice were treated with either uric acid (intraperitoneally injection 250 mg/kg) or saline for a total of 13 d. We showed that uric acid improved behavioral performances and cognition of PD mice, increased TH-positive dopaminergic neurons and decreased GFAP-positive astrocytes in substantia nigra (SN). Uric acid increased mRNA and protein expressions of Nrf2 and three Nrf2-responsive genes, including γ-glutamate-cysteine ligase catalytic subunit (γ-GCLC), heme oxygenase-1 (HO-1) and NQO1. Uric acid significantly increased superoxide dismutase (SOD), CAT, glutathione (GSH) levels and decreased malondialdehyde (MDA) level in SN regions of MPTP-treated mice. Uric acid inhibited the hippocampal expression of IL-1ß and decreased serum and hippocampus levels of interleukin-1ß (IL-1ß), IL-6 and tumor necrosis factor-α (TNF-α). In conclusion, uric acid demonstrates neuroprotective properties for dopaminergic neurons in PD mice through modulation of neuroinflammation and oxidative stress.


Subject(s)
Antioxidant Response Elements/drug effects , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology , Uric Acid/pharmacology , Animals , Behavior, Animal/drug effects , Cognition/drug effects , Cytokines/blood , Cytokines/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Hippocampus/physiopathology , Inflammation/drug therapy , Inflammation/pathology , MPTP Poisoning/drug therapy , MPTP Poisoning/physiopathology , MPTP Poisoning/psychology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Parkinson Disease/psychology , Signal Transduction/drug effects
13.
Brain Res Bull ; 132: 129-138, 2017 06.
Article in English | MEDLINE | ID: mdl-28576659

ABSTRACT

Hyperactivity of the glutamatergic system is involved in excitotoxicity and neurodegeneration in Parkinson's disease (PD) so that glutamatergic modulation maybe a potential therapeutic target for PD. Ceftriaxone (CEF) has been reported to increase glutamate uptake by increasing glutamate transporter expression and has been demonstrated neuroprotective effects in animal study. The aim of this study was to determine the effects of CEF on behavior and neurogenesis in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD rat model. MPTP was stereotaxically injected into the substantia nigra pars compacta (SNc) of male Wistar rats. Starting on the same day after MPTP lesioning (day 0), the rats were injected daily with either CEF or saline for 14days and underwent a T-maze test on days 8-10 and an object recognition test on days 12-14, then the brain was taken for histological evaluation on day 15. The results showed that MPTP lesioning resulted in decreased motor function, working memory, and object recognition and reduced neurogenesis in the substantial nigra and dentate gyrus of the hippocampus. These behavioral and neuronal changes were prevented by CEF treatment. To our knowledge, this is the first study showing that CEF prevents loss of neurogenesis in the brain of PD rats. CEF may therefore have clinical potential in the treatment of PD.


Subject(s)
Antiparkinson Agents/pharmacology , Ceftriaxone/pharmacology , MPTP Poisoning/drug therapy , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Nootropic Agents/pharmacology , Animals , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Dentate Gyrus/physiopathology , MPTP Poisoning/pathology , MPTP Poisoning/physiopathology , MPTP Poisoning/psychology , Male , Maze Learning/drug effects , Memory, Short-Term/drug effects , Motor Activity/drug effects , Neurons/drug effects , Neurons/pathology , Neurons/physiology , Rats, Wistar , Recognition, Psychology/drug effects , Substantia Nigra/drug effects , Substantia Nigra/pathology , Substantia Nigra/physiopathology
14.
Inflammopharmacology ; 25(4): 471-484, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28577132

ABSTRACT

Our aim was to investigate whether tangeretin, a citrus flavonoid, was able to prevent neuroinflammation and improve dementia in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced rodent model of Parkinson's disease (PD). MPTP-HCl was infused into the substantia nigra pars compacta of male Sprague-Dawley rats. Tangeretin (50, 100 or 200 mg/kg body weight) was administered orally starting 3 days prior to MPTP injection and was continued for 20 days following injection. MPTP-lesioned rats revealed motor dysfunction in bar test and rota rod tests. Deficits in working memory and object recognition function were also observed following MPTP induction. Tangeretin treatment significantly attenuated the memory deficits and improved motor functions and cognition. Immunohistochemical analysis reveals the protective effects of tangeretin against MPTP lesion-induced dopaminergic degeneration and hippocampal neuronal loss. Tangeretin reduced expression of inflammatory mediators-COX-2, iNOS-as well reduced the levels of cytokines-interleukins (IL)-IL-1ß, IL-6 and IL-2. The experimental data suggest tangeretin as an effective candidate drug with potential for prevention and treatment of neuroinflammation and dementia associated with PD.


Subject(s)
Dementia/drug therapy , Flavones/therapeutic use , Inflammation Mediators/antagonists & inhibitors , MPTP Poisoning/drug therapy , Maze Learning/drug effects , Animals , Cognition/drug effects , Cognition/physiology , Dementia/metabolism , Dementia/psychology , Dose-Response Relationship, Drug , Flavones/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/psychology , Inflammation Mediators/metabolism , MPTP Poisoning/metabolism , MPTP Poisoning/psychology , Male , Maze Learning/physiology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/psychology , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats , Rats, Sprague-Dawley
15.
J Pharmacol Sci ; 134(1): 55-58, 2017 May.
Article in English | MEDLINE | ID: mdl-28456375

ABSTRACT

Cognitive impairment often occurs in Parkinson's disease (PD), but the mechanism of onset remains unknown. Recently, we reported that PD model mice produced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) show facilitation of hippocampal memory extinction, which may be the cause of cognitive impairment in PD. When we examined the cAMP/CREB signaling in the hippocampus, decreased levels of cAMP and phosphorylated CREB were observed in the dentate gyrus (DG) of MPTP-treated mice. Administration of rolipram improved the memory deficits with concomitant recovery of cAMP and phosphorylated CREB levels, suggesting that reduced cAMP/CREB signaling in the DG leads to cognitive impairment in MPTP-treated mice.


Subject(s)
Fear , Hippocampus/metabolism , MPTP Poisoning/drug therapy , Memory/drug effects , Parkinson Disease/drug therapy , Rolipram/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Behavior, Animal/drug effects , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Extinction, Psychological , Hippocampus/drug effects , MPTP Poisoning/metabolism , MPTP Poisoning/psychology , Male , Mice , Mice, Inbred C57BL , Parkinson Disease/metabolism , Parkinson Disease/psychology
16.
Behav Brain Res ; 325(Pt A): 51-62, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28219749

ABSTRACT

Parkinson's disease is a chronic neurodegenerative disorder with the core motor features of resting tremor, bradykinesia, rigidity, and postural instability. Non-motor symptoms also occur, and include cognitive dysfunction, mood disorders, anosmia (loss of smell), and REM sleep disturbances. As the development of medications and other therapies for treatment of non-motor symptoms is ongoing, it is essential to have animal models that aid in understanding the neural changes underlying non-motor PD symptoms and serve as a testing ground for potential therapeutics. We investigated several non-motor symptoms in 10 adult male marmosets using the MPTP model, with both the full (n=5) and partial (n=5) MPTP dosing regimens. Baseline data in numerous domains were collected prior to dosing; assessments in these same domains occurred post-dosing for 12 weeks. Marmosets given the partial MPTP dose (designed to mimic the early stages of the disease) differed significantly from marmosets given the full MPTP dose in several ways, including behavior, olfactory discrimination, cognitive performance, and social responses. Importantly, while spontaneous recovery of PD motor symptoms has been previously reported in studies of MPTP monkeys and cats, we did not observe recovery of any non-motor symptoms. This suggests that the neurochemical mechanisms behind the non-motor symptoms of PD, which appear years before the onset of symptoms, are independent of the striatal dopaminergic transmission. We demonstrate the value of assessing a broad range of behavioral change to detect non-motor impairment, anosmia, and differences in socially appropriate responses, in the marmoset MPTP model of early PD.


Subject(s)
Parkinsonian Disorders/psychology , Psychomotor Performance/drug effects , Substantia Nigra/drug effects , Substantia Nigra/pathology , Animals , Behavior, Animal/drug effects , Callithrix , Executive Function/drug effects , MPTP Poisoning/pathology , MPTP Poisoning/psychology , Male , Motor Activity/drug effects , Phenotype , Smell/drug effects , Social Behavior
17.
Neuropharmacology ; 110(Pt A): 48-58, 2016 11.
Article in English | MEDLINE | ID: mdl-27424102

ABSTRACT

Istradefylline (KW-6002), an adenosine A2A receptor antagonist, is used adjunct with optimal doses of L-3,4-dihydroxyphenylalanine (l-DOPA) to extend on-time in Parkinson's disease (PD) patients experiencing motor fluctuations. Clinical application of istradefylline for the management of other l-DOPA-induced complications, both motor and non-motor related (i.e. dyskinesia and cognitive impairments), remains to be determined. In this study, acute effects of istradefylline (60-100 mg/kg) alone, or with optimal and sub-optimal doses of l-DOPA, were evaluated in two monkey models of PD (i) the gold-standard 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated macaque model of parkinsonian and dyskinetic motor symptoms and (ii) the chronic low dose (CLD) MPTP-treated macaque model of cognitive (working memory and attentional) deficits. Behavioural analyses in l-DOPA-primed MPTP-treated macaques showed that istradefylline alone specifically alleviated postural deficits. When combined with an optimal l-DOPA treatment dose, istradefylline increased on-time, enhanced therapeutic effects on bradykinesia and locomotion, but exacerbated dyskinesia. Istradefylline treatment at specific doses with sub-optimal l-DOPA specifically alleviated bradykinesia. Cognitive assessments in CLD MPTP-treated macaques showed that the attentional and working memory deficits caused by l-DOPA were lowered after istradefylline administration. Taken together, these data support a broader clinical use of istradefylline as an adjunct treatment in PD, where specific treatment combinations can be utilised to manage various l-DOPA-induced complications, which importantly, maintain a desired anti-parkinsonian response.


Subject(s)
Cognition Disorders/drug therapy , Dyskinesia, Drug-Induced/drug therapy , Levodopa/administration & dosage , MPTP Poisoning/drug therapy , Purines/administration & dosage , Adenosine A2 Receptor Antagonists/administration & dosage , Animals , Cognition Disorders/physiopathology , Cognition Disorders/psychology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Drug Therapy, Combination , Dyskinesia, Drug-Induced/physiopathology , Dyskinesia, Drug-Induced/psychology , Female , Hypokinesia/drug therapy , Hypokinesia/physiopathology , Hypokinesia/psychology , Levodopa/toxicity , MPTP Poisoning/physiopathology , MPTP Poisoning/psychology , Macaca fascicularis , Motor Skills Disorders/drug therapy , Motor Skills Disorders/physiopathology , Motor Skills Disorders/psychology , Treatment Outcome
18.
Exp Neurol ; 279: 187-196, 2016 May.
Article in English | MEDLINE | ID: mdl-26946223

ABSTRACT

Neurophysiological changes in the basal ganglia thalamo-cortical circuit associated with the development of parkinsonian motor signs remain poorly understood. Theoretical models have ranged from those emphasizing changes in mean discharge rate to increased oscillatory activity within the beta range. The present study characterized neuronal activity within and across the internal and external segments of the globus pallidus as a function of motor severity using a staged, progressively severe 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinsonism in three rhesus monkeys. An increase in coherence between neuronal pairs across the external and internal globus pallidus was present in multiple frequency bands in the parkinsonian state; both the peak frequency of oscillatory coherence and the variability were reduced in the parkinsonian state. The incidence of 8-20Hz oscillatory activity in the internal globus pallidus increased with the progression of the disease when pooling the data across the three animals; however it did not correlate with motor severity when assessed individually and increased progressively in only one of three animals. No systematic relationship between mean discharge rates or the incidence or structure of bursting activity and motor severity was observed. These data suggest that exaggerated coupling across pallidal segments contribute to the development of the parkinsonian state by inducing an exaggerated level of synchrony and loss of focusing within the basal ganglia. These data further point to the lack of a defined relationship between rate changes, the mere presence of oscillatory activity in the beta range and bursting activity in the basal ganglia to the motor signs of Parkinson's disease.


Subject(s)
Globus Pallidus/physiopathology , MPTP Poisoning/physiopathology , Parkinson Disease/physiopathology , Animals , Behavior, Animal , Beta Rhythm , Disease Models, Animal , Disease Progression , Electroencephalography , Female , Globus Pallidus/pathology , MPTP Poisoning/pathology , MPTP Poisoning/psychology , Macaca mulatta , Male , Movement Disorders/etiology , Movement Disorders/physiopathology , Neurons/pathology , Parkinson Disease/pathology , Parkinson Disease/psychology
19.
Behav Brain Res ; 298(Pt B): 192-201, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26393431

ABSTRACT

In Parkinson's disease, cognitive deficits manifest as fronto-striatally-mediated executive dysfunction, with impaired attention, planning, judgment, and impulse control. We examined changes in executive function in mice lesioned with subchronic 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) using a 3-choice serial reaction-time (SRT) task, which included measures of sustained attention and impulse control. Each trial of the baseline SRT task comprised a pseudo-random pre-cue period ranging from 3 to 8 s, followed by a 1-s cue duration. MPTP impaired all measures of impulsive behavior acutely, but with additional training their performance normalized to saline control levels. When challenged with shorter cue durations, MPTP-lesioned mice had significantly slower reaction times than wild-type mice. When challenged with longer pre-cue times, the MPTP-lesioned mice exhibited a loss of impulse control at the longer durations. In lesioned mice, striatal dopamine was depleted by 54% and the number of tyrosine-hydroxylase-positive neurons in the substantia nigra pars compacta was reduced by 75%. Serotonin (5-HT) was unchanged in the striatum and prefrontal cortex (PFC), but the ratio of 5-hydroxyindolacetic acid (5-HIAA) to 5-HT was significantly reduced in the MPTP group in the PFC. In lesioned mice, prefrontal 5-HIAA/5-HT was significantly correlated with the executive impairments and striatal norepinephrine was associated with slower reaction times. None of the neurochemical measures was significantly associated with behavior in saline-treated controls. Taken together, these results show that prefrontal 5-HT turnover may play a pivotal role in MPTP-induced executive dysfunction.


Subject(s)
Executive Function/physiology , MPTP Poisoning/physiopathology , MPTP Poisoning/psychology , Prefrontal Cortex/physiopathology , Serotonin/metabolism , Animals , Attention/physiology , Corpus Striatum/physiopathology , Disease Models, Animal , Dopamine/metabolism , Hydroxyindoleacetic Acid/metabolism , Impulsive Behavior/physiology , Male , Mice, Inbred C57BL , Neurons/physiology , Neuropsychological Tests , Pars Compacta/physiopathology , Reaction Time/physiology , Tyrosine 3-Monooxygenase/metabolism
20.
Neurotox Res ; 28(3): 222-31, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26156429

ABSTRACT

Considering that depression is a common non-motor comorbidity of Parkinson's disease and that agmatine is an endogenous neuromodulator that emerges as a potential agent to manage diverse central nervous system disorders, this study investigated the antidepressant-like effect of agmatine in mice intracerebroventricularly (i.c.v.) injected with the dopaminergic neurotoxin 1-methyl-4-phenylpyridinium (MPP(+)). Male C57BL6 mice were treated with agmatine (0.0001, 0.1 or 1 mg/kg) and 60 min later the animals received an i.c.v. injection of MPP(+) (1.8 µg/site). Twenty-four hours after MPP(+) administration, immobility time, anhedonic behavior, and locomotor activity were evaluated in the tail suspension test (TST), splash test, and open field test, respectively. Using Western blot analysis, we investigated the putative modulation of MPP(+) and agmatine on striatal and frontal cortex levels of tyrosine hydroxylase (TH) and brain-derived neurotrophic factor (BDNF). MPP(+) increased the immobility time of mice in the TST, as well as induced an anhedonic-like behavior in the splash test, effects which were prevented by pre-treatment with agmatine at the three tested doses. Neither drug, alone or in combination, altered the locomotor activity of mice. I.c.v. administration of MPP(+) increased the striatal immunocontent of TH, an effect prevented by the three tested doses of agmatine. MPP(+) and agmatine did not alter the immunocontent of BDNF in striatum and frontal cortex. These results demonstrate for the first time the antidepressant-like effects of agmatine in an animal model of depressive-like behavior induced by the dopaminergic neurotoxin MPP(+).


Subject(s)
Agmatine/pharmacology , Antidepressive Agents/pharmacology , Depressive Disorder/drug therapy , Depressive Disorder/physiopathology , MPTP Poisoning/physiopathology , Anhedonia/drug effects , Anhedonia/physiology , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/drug effects , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Depressive Disorder/etiology , Depressive Disorder/pathology , Dose-Response Relationship, Drug , Frontal Lobe/drug effects , Frontal Lobe/pathology , Frontal Lobe/physiopathology , MPTP Poisoning/complications , MPTP Poisoning/drug therapy , MPTP Poisoning/psychology , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...