Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.561
Filter
1.
Neurochem Int ; 151: 105215, 2021 12.
Article in English | MEDLINE | ID: mdl-34710535

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease characterized by progressive loss of the nigrostriatal dopaminergic neurons that are associated with motor alterations and non-motor manifestations (such as depression). Neuroinflammation is a process with a critical role in the pathogenesis of PD. In this regard, toll-like receptor 4 (TLR4) is a central mediator of immune response in PD. Moreover, there are gender-related differences in the incidence, prevalence, and clinical features of PD. Therefore, we aimed to elucidate the role of TLR4 in the sex-dependent response to dopaminergic denervation induced by 6-hydroxydopamine (6-OHDA) in mice. Female and male adult wildtype (WT) and TLR4 knockout (TLR4-/-) mice were administered with unilateral injection of 6-OHDA in the dorsal striatum, and non-motor and motor impairments were evaluated for 30 days, followed by biochemistry analysis in the substantia nigra pars compacta (SNc), dorsal striatum, and dorsoventral cortex. Early non-motor impairments (i.e., depressive-like behavior and spatial learning deficits) induced by 6-OHDA were observed in the male WT mice but not in male TLR4-/- or female mice. Motor alterations were observed after administration of 6-OHDA in both strains, and the lack of TLR4 was also related to motor commitment. Moreover, ablation of TLR4 prevented 6-OHDA-induced dopaminergic denervation and microgliosis in the SNc, selectively in female mice. These results reinforced the existence of sex-biased alterations in PD and indicated TLR4 as a promising therapeutic target for the motor and non-motor symptoms of PD, which will help counteract the neuroinflammatory and neurodegenerative processes.


Subject(s)
Brain/drug effects , Parkinson Disease/drug therapy , Sex Factors , Toll-Like Receptor 4/metabolism , Animals , Brain/pathology , Disease Models, Animal , Female , Hydroxydopamines/pharmacology , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Nerve Degeneration/chemically induced , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/pathology , Parkinson Disease/genetics , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/genetics
2.
J Pharm Pharmacol ; 71(8): 1271-1281, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31144300

ABSTRACT

OBJECTIVE: To investigate the effect of desferrioxamine (DFO) and dextromethorphan (DXM) combination in animal model of Parkinson's disease (PD). METHODS: The PD was induced in rats through intracerebroventricular administration of 6-hydroxydopamine (6-OHDA) using stereotaxic apparatus. The animals were subjected to behavioural assessments and neurobiochemicals estimation followed by immunohistochemistry staining of neuron specific enolase (NSE) in striatum. KEY FINDINGS: Desferrioxamine and DXM combination has significantly reversed the catalepsy behaviour and elevated the antioxidant enzymes (SOD, CAT, GSH) and dopamine levels. Interestingly, the level of glutamate, nitric oxide, cytokines (IL-1ß, TNF-α) and NSE expressions were found to be decreased in striatum region of 6-OHDA-administered rats. The combination of DFO and DXM has shown synergism in most of the parameters studied, when compared to per se treatment. CONCLUSIONS: The reversal of catalepsy behaviour represents the protective effect of above combination on dopamine neurons in striatum from 6-OHDA toxicity. The mechanism of DFO and DXM combination might be attributed through attenuation of glutamate-induced excitotoxicity in neurons through ameliorating the reactive oxygen species and pro-inflammatory cytokines release. Treatment with DFO and DXM combination could control the multiple events in the pathogenesis of PD.


Subject(s)
Catalepsy/drug therapy , Corpus Striatum/drug effects , Deferoxamine/pharmacology , Dextromethorphan/pharmacology , Glutamic Acid/metabolism , Hydroxydopamines/pharmacology , Animals , Antioxidants/pharmacology , Catalepsy/metabolism , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Drug Combinations , Drug Synergism , Male , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Rats , Rats, Wistar
3.
Neurosci Lett ; 687: 177-182, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30266672

ABSTRACT

INTRODUCTION: Parkinson's Disease (PD) is a progressive neurodegenerative disorder, hallmark of which is loss of nigral dopaminergic neurons. Since a Hypericum polyanthemum extract inhibits monoamine reuptake and some of its constituents present cytotoxic properties, the aim of this study was to evaluate the effect of this extract in an animal PD model. METHODS: Adult Wistar rats (110 days old) received 6-hydroxydopamine (6-OHDA) infusions into the right medial forebrain bundle. A cyclohexane extract from aerial parts of H. polyanthemum (POL; 90 mg/kg/administration; gavage) was administered in three different regimens. In Regimens 1 and 2, rats received 3 administrations of POL starting 4 or 24 h after 6-OHDA infusion, respectively. In Regimen 3, these administrations were carried out 1 day before any evaluation of ipsilateral rotational activity induced by methylphenidate (MP, 20 mg/kg, i.p.). MP was administered 10, 45, and 85 days after 6-OHDA infusion in all groups. Nigral tyrosine hydroxylase (TH) immunocontent was evaluated 120 days after 6-OHDA infusion in animals submitted to Regimen 2 only. The effect of POL on apomorphine-induced climbing behavior in non-lesioned adult CF1 mice (60 days old) treated with POL was also evaluated. RESULTS: Regimen 2 increased MP-induced rotational activity and decreased nigral TH levels in 6-OHDA-lesioned rats. Rotational activity was not altered in regimens 1 and 3. In addition, no change in climbing behavior was observed in non-lesioned mice. CONCLUSION: Together, these results indicate that, in 6-OHDA-lesioned rats, a cyclohexane H. polyanthemum extract potentiates neurotoxicity and MP-induced motor asymmetry depending on the time of administration. In the short term, it seems to not act directly on mice dopaminergic receptors.


Subject(s)
Behavior, Animal/drug effects , Hydroxydopamines/pharmacology , Hypericum/metabolism , Motor Activity/drug effects , Animals , Disease Models, Animal , Dopamine/pharmacology , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/drug therapy , Rats, Wistar , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism
4.
Pharmazie ; 71(8): 465-471, 2016 08 01.
Article in English | MEDLINE | ID: mdl-29442034

ABSTRACT

Parkinson's disease (PD) is a degenerative brain disorder characterized by motor symptoms and loss of dopaminergic (DA) neurons in the substantia nigra. The mechanisms for DA cell death in PD have been extensively investigated using PC12 cells treated with a dopamine neurotoxin 6-hydroxydopamine (6-OHDA). 6-OHDA may induce both autophagy and apoptosis in PC12 cells. However, it remains unclear whether crosstalk occurs between autophagy and apoptosis in PC12 cells treated with 6-OHDA and whether Raf-1/ERK1/2 and their phosphorylation status play a role in autophagy. In this study, we used MDC staining assay and flow cytometry and found that 6-OHDA induced autophagy in PC12 cells. This induction was inhibited by the autophagy inhibitor 3-MA. Our electron microscopy observations also supported 6-OHDA induced autophagy in PC12 cells. Apoptosis of PC12 cells was increased with inhibition of autophagy by 3-MA. In addition, Inhibition of Raf-1 resulted in a decreased 6-OHDA-induced autophagy rate among PC12 cells. Phosphorylation levels of Raf-1 and ERK1/2 were increased in PC12 cells treated with 6-OHDA and inhibited by co-treatment with 6-OHDA and 3-MA. These data suggest that crosstalk between 6-OHDA-induced apoptosis and autophagy in PC12 cells may be regulated via the Raf-1/ERK1/2 signaling pathway. Our data suggest a mechanism for 6-OHDA toxicity in PC12 cells, contributing to our understanding of the pathogenesis of PD.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Hydroxydopamines/pharmacology , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-raf/metabolism , Receptor Cross-Talk/drug effects , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Cell Survival/drug effects , PC12 Cells , Proto-Oncogene Proteins c-raf/drug effects , Rats
5.
Ross Fiziol Zh Im I M Sechenova ; 101(6): 721-30, 2015 Jun.
Article in Russian | MEDLINE | ID: mdl-26470491

ABSTRACT

The study presents a comprehensive of the metabolism and the fractional composition of li- pids surfactant, water balance, hemostatic activity of the lungs in neyrodegeneration substantia nigra of the brain induced by stereotaxic microinjection of 6--hydroxydopamine and systemic administration of haloperidol. It is shown that a breach of dopaminergic neurotransmission leads to deterioration of surface-active properties of the alveolar lining of the complex against a decrea- se of phospholipids, cholesterol, phosphatidylcholine and lysophospholipids enhance the pulmo- nary surfactant in the activation of phospholipase hydrolysis and lipid peroxidation. Intranigral introduction neurotoxin accompanied by increased blood supply to the lungs and the blood coagu- lation potential of the pulmonary circulation, the blockade D2-receptors--hyporhydration lung tissue. The results obtained indicate the formation of dysregulation pneumopathy dysfunction nigrostriatal dopaminergic system.


Subject(s)
Dopamine Antagonists/adverse effects , Haloperidol/adverse effects , Hydroxydopamines/adverse effects , Lung , Substantia Nigra , Water-Electrolyte Balance/drug effects , Animals , Dopamine Antagonists/pharmacology , Haloperidol/pharmacology , Hydroxydopamines/pharmacology , Lung/metabolism , Lung/pathology , Lung/physiopathology , Male , Rats , Substantia Nigra/metabolism , Substantia Nigra/pathology , Substantia Nigra/physiopathology , Synaptic Transmission/drug effects
6.
Neurochem Int ; 64: 18-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24220540

ABSTRACT

Oxidative stress-induced neuronal death plays a pivotal role in pathogenesis of neurodegenerative disorders. Recently, 7,8-dihydroxyflavone (7,8-DHF) has been shown to exert neuroprotective effects by acting as a selective tyrosine kinase receptor B (TrkB) agonist. In addition, the antioxidant action of 7,8-DHF may protect neuronal cells against oxidative injury. In the present study, we used PC12 cells, a cell line generally thought to lack TrkB, to investigate the effect of 7,8-DHF on 6-hydroxydopamine (6-OHDA)-induced cytotoxicity and the underlying mechanism. We found that 7,8-DHF effectively prevented cell death, apoptosis and mitochondrial dysfunction induced by 6-OHDA. In a cell free system, 7,8-DHF did not slow down extracellular auto-oxidation of 6-OHDA which may generate H2O2. However, We found that 7,8-DHF dramatically reduced cellular malondialdehyde content and phospho-histone H2A.X protein level. 7,8-DHF also elevated total superoxide dismutase activity in 6-OHDA-treated cells. These results indicate that 7,8-DHF might protect PC12 cells against 6-OHDA-induced cytotoxicity through its powerful antioxidant activity. By acting as a potent TrkB agonist and an antioxidant together with its easiness to pass across blood-brain barrier, 7,8-DHF may be developed into a promising candidate in treatment of neurodegenerative diseases.


Subject(s)
Antioxidants/pharmacology , Flavones/pharmacology , Hydroxydopamines/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Cell Death/drug effects , Cell Line , Hydrogen Peroxide/pharmacology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/metabolism , Oxidative Stress/drug effects , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
7.
Int J Neurosci ; 123(10): 705-10, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23590664

ABSTRACT

Glial-cell-line-derived neurotrophic factor (GDNF) has been shown to protect dopaminergic (DA) neurons against 6-hydroxydopamine (6-OHDA) toxicity. The mechanism underlying the antiapoptosis role of GDNF still needs further studies. We previously observed that nuclear factor-kappaB (NF-κB) signaling pathway, i.e. p65/p52, mediated the antiapoptosis role of GDNF in MN9D cells. Here, the DA cell line MN9D was used to explore the mechanisms underlying NF-κB p65/p52-mediated protection role of GDNF in DA neurons. The results showed that GDNF pretreatment blocked the apoptotic effects induced by 6-OHDA, with the upregulation of the antiapoptotic protein, Bcl-2 and Bcl-w, as well as the downregulation of the proapoptotic proteins, Bax and Bad. Furthermore, when sip100 plasmids were transfected into MN9D cells to inhibit the expression of p100, which was the precursor of p52, the effects of GDNF on upregulating Bcl-2 and Bcl-w were attenuated. These results indicated that GDNF could protect MN9D cells from apoptosis induced by 6-OHDA via upregulating Bcl-2 and Bcl-w expressions and downregulating Bax and Bad expressions. Moreover, NF-κB p65/p52 signaling mediated the effects of GDNF on Bcl-2 and Bcl-w expressions.


Subject(s)
Apoptosis/drug effects , Dopaminergic Neurons/metabolism , Genes, bcl-2/genetics , Glial Cell Line-Derived Neurotrophic Factor/genetics , Hydroxydopamines/pharmacology , NF-kappa B p52 Subunit/genetics , Proteins/genetics , Transcription Factor RelA/genetics , Animals , Apoptosis Regulatory Proteins , Blotting, Western , Dopaminergic Neurons/drug effects , Mesencephalon/cytology , Mice , NF-kappa B p52 Subunit/physiology , Plasmids/genetics , RNA, Small Interfering/genetics , Transcription Factor RelA/physiology , Transfection , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism
8.
Neuropeptides ; 47(3): 139-47, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23490004

ABSTRACT

The hypothalamic neurochemicals neuropeptide Y (NPY), orexin-A (ORX), and oxytocin (OXY) exert glucoregulatory effects upon intracerebral administration, findings that support their potential function within neural pathways that maintain glucostasis. Current understanding of how these neurotransmitter systems respond to the diabetes mellitus complication, insulin-induced hypoglycemia, is limited to knowledge of neuropeptide gene transcriptional reactivity. We investigated the hypothesis that hypoglycemia elicits hypothalamic site-specific alterations in levels of these neurochemicals, and that adjustments in local neurotransmitter availability may be regulated by catecholaminergic (CA) input from the caudal dorsomedial hindbrain. The arcuate (ARH) and paraventricular (PVH) hypothalamic nuclei and lateral hypothalamic area (LHA) were each microdissected from adult male rats pretreated by caudal fourth ventricular administration of the selective CA neurotoxin, 6-hydroxydopamine (6-OHDA), or vehicle prior to insulin (INS)-induced hypoglycemia. Hypoglycemia stimulated ARH NPY gene expression and NPY accumulation in the ARH and LHA, but not PVH. 6-OHDA pretreatment did not modify the positive NPY mRNA response to INS, but blunted hypoglycemic augmentation of ARH and LHA NPY content while increasing PVH NPY levels in response to hypoglycemia. INS-treated rats exhibited diminished LHA ORX gene expression and increased [ARH; LHA] or decreased [PVH] tissue ORX protein levels. 6-OHDA+INS animals showed a comparable decline in ORX transcripts, but attenuated augmentation of ARH and LHA ORX content and elevated PVH ORX levels. OT mRNA and protein were respectively decreased or unchanged during hypoglycemia, responses that were uninfluenced by hindbrain CA nerve cell destruction. These results illustrate divergent adjustments in glucoregulatory neurotransmitter gene expression and site-specific protein accumulation in the hypothalamus during hypoglycemia. Evidence that 6-OHDA pretreatment does not modify NPY or ORX transcriptional reactivity to hypoglycemia, but alters hypoglycemic patterns of NPY and ORX accretion implicates dorsomedial hindbrain CA neurons in regulation of translation/post-translational processing and site-specific availability of these neurotransmitters in the hypothalamus during hypoglycemia.


Subject(s)
Catecholamines/metabolism , Glucose/metabolism , Hypoglycemia/metabolism , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Neuropeptide Y/biosynthesis , Neuropeptides/biosynthesis , Oxytocin/biosynthesis , Rhombencephalon/metabolism , Animals , Blood Glucose/metabolism , Blotting, Western , Hydroxydopamines/pharmacology , Hypoglycemia/chemically induced , Hypoglycemic Agents , Immunohistochemistry , Insulin , Intracellular Signaling Peptides and Proteins/genetics , Male , Neuropeptide Y/genetics , Neuropeptides/genetics , Neurotoxins/metabolism , Neurotransmitter Agents/metabolism , Orexins , Oxytocin/genetics , Polymerase Chain Reaction , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Sympathectomy, Chemical
9.
Anat Rec (Hoboken) ; 294(5): 875-82, 2011 May.
Article in English | MEDLINE | ID: mdl-21455956

ABSTRACT

Sympathetic regulation plays an important role in fetal survival and development during early pregnancy. Maternal adaptations to pregnancy may involve changes of the spleen in its structure, size, and function. This study was therefore designed to investigate the effects of sympathetic nerves on these adaptations in the maternal spleen of mice during early pregnancy. The adult female mice were intraperitoneally injected with neurotoxin 6-hydroxydopamine (6-OHDA) for 5 consecutive days to selectively destroy the sympathetic nerves. The results were as follows: (1) the splenic weight was reduced in the 6-OHDA group by 10.9%-13.0% at E5-E9 (P < 0.05) when compared with the control group. (2) The splenic nodule and periarterial lymphatic sheath of the 6-OHDA-treated mice were smaller than those of control mice. The proliferating cell nuclear antigen positive cells of 6-OHDA-treated group were decreased by 10.9%-16.2% (P < 0.05) at E1-E9. (3) Lymphocyte proliferation indices in response to concanavalin A or lipopolysaccharide were significantly decreased (P < 0.05) in the 6-OHDA group. (4) When compared with control mice, the superoxide dismutase and glutathione peroxidase activities of 6-OHDA-treated mice were decreased by 14.3%-21.9% (P < 0.05) at E1-E9 and 17.4%-25.0% (P < 0.05) at E3-E9, respectively. In contrast, the malondialdehyde content of 6-OHDA group was increased by 10.6%-38.6% (P < 0.05) at E3-E9. The results demonstrated the regulation of pregnancy-dependent adaptations in the spleen through the sympathetic nerve activity.


Subject(s)
Antioxidants/pharmacology , Lymphocytes/cytology , Prenatal Exposure Delayed Effects , Spleen/cytology , Sympathetic Nervous System/physiology , Animals , Concanavalin A/pharmacology , Female , Glutathione Peroxidase/metabolism , Hydroxydopamines/pharmacology , Lipid Peroxidation/drug effects , Lipopolysaccharides/pharmacology , Lymphocyte Activation , Lymphocytes/drug effects , Lymphocytes/metabolism , Male , Malondialdehyde/metabolism , Mice , Organ Size/drug effects , Oxidants/pharmacology , Pregnancy , Pregnancy, Animal , Proliferating Cell Nuclear Antigen/metabolism , Spleen/metabolism
10.
Toxicology ; 283(2-3): 118-28, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21397656

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative movement disorder characterized by selective loss of dopaminergic neurons in the substantia nigra. 6-Hydroxydopamine (6-OHDA) is a catecholaminergic neurotoxin widely used to produce experimental models of PD and has been reported to cause oxidative and/or nitrosative stress. In this study, we have investigated 6-OHDA-induced nitrosative cell death and its self-defense mechanism in C6 glioma cells. Treatment of C6 cells with 6-OHDA increased expression of inducible nitric oxide synthase (iNOS) and subsequent production of nitric oxide (NO). Furthermore 6-OHDA treatment led to peroxynitrite generation and nitrotyrosine formation. 6-OHDA-induced nitrosative stress ultimately caused apoptotic cell death as determined by decreased Bcl-2/Bax ratio, activation of c-Jun N-terminal kinase (JNK), and cleavage of caspase-3 and poly(ADP-ribose)polymerase (PARP), which were attenuated by peroxynitrite decomposition catalyst, 5,10,15,20-tetrakis(4-sulfonatophenyl)prophyrinato iron(III) (FeTPPS). In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. To elucidate the molecular mechanism underlying 6-OHDA-mediated HO-1 induction, we have examined the possible involvement of NF-E2-related factor 2 (Nrf2), which plays an important role in the transcriptional regulation of phase II detoxifying and antioxidant enzymes. 6-OHDA treatment increased nuclear translocation and transcriptional activity of Nrf2, which seemed to be partly mediated by activation of upstream kinases such as Akt/protein kinase B (PKB). Taken together these findings suggest that HO-1 up-regulation via Nrf2 activation may mediate the cellular adaptive survival response to 6-OHDA-induced nitrosative cell death in C6 glioma cells.


Subject(s)
Antioxidants/metabolism , Cell Survival/drug effects , Reactive Nitrogen Species/metabolism , Blotting, Western , Coloring Agents , Glioma/pathology , Heme Oxygenase-1/metabolism , Humans , Hydroxydopamines/pharmacology , Immunohistochemistry , In Situ Nick-End Labeling , Luciferases/biosynthesis , Luciferases/genetics , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Nerve Tissue Proteins/metabolism , Nitric Oxide/metabolism , Porphyrins/pharmacology , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
11.
Prog Neuropsychopharmacol Biol Psychiatry ; 34(6): 877-81, 2010 Aug 16.
Article in English | MEDLINE | ID: mdl-20398719

ABSTRACT

Previous studies have revealed that 5-HT(1A) agonists ameliorate antipsychotic-induced extrapyramidal symptoms (EPS) through postsynaptic 5-HT(1A) receptors. Here, we conducted an intracerebral microinjection study of (+/-)-8-hydroxy-2-(di-n-propylamino)-tetralin ((+/-)8-OH-DPAT) to determine the action site of the 5-HT(1A) agonist in alleviating EPS. Bilateral microinjection of(+/-)8-OH-DPAT (5 microg/1microL per side) either into the primary motor cortex (MC) or the dorsolateral striatum (dlST) significantly attenuated haloperidol-induced catalepsy in rats. The anticataleptic action of (+/-)8-OH-DPAT was more prominent with the MC injection than with the dlST injection. WAY-100135 (a selective 5-HT(1A) antagonist) completely antagonized the reversal of haloperidol-induced catalepsy both by intracortical and intrastriatal (+/-)8-OH-DPAT. Furthermore, lesioning of dopamine neurons with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (30 mg/kg/day, i.p., for 4 days) did not alter the anti-EPS actions of (+/-)8-OH-DPAT in a mouse pole test. The present results strongly suggest that 5-HT(1A) agonist alleviates antipsychotic-induced EPS by activating postsynaptic 5-HT(1A) receptors in the MC and dlST, probably through non-dopaminergic mechanisms.


Subject(s)
Basal Ganglia Diseases/metabolism , Corpus Striatum/metabolism , Haloperidol/pharmacology , Motor Cortex/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , Animals , Antipsychotic Agents/pharmacology , Basal Ganglia Diseases/chemically induced , Corpus Striatum/drug effects , Hydroxydopamines/pharmacology , Male , Microinjections , Motor Cortex/drug effects , Neurons/drug effects , Neurons/metabolism , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin Antagonists/pharmacology , Statistics, Nonparametric
12.
Mol Ther ; 16(8): 1392-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18545225

ABSTRACT

In Parkinson's disease (PD) chronic inflammation occurs in the substantia nigra (SNc) concurrently with dopaminergic neurodegeneration. In models of PD, microglial activation precedes neurodegeneration in the SNc, suggesting that the underlying pathogenesis involves a complex response in the nigrostriatal pathway, and that the innate immune system plays a significant role. We have investigated the neuroprotective effect of an adeno-associated viral type-2 (AAV2) vector containing the complementary DNA (cDNA) for human interleukin-10 (hIL-10) in the unilateral 6-hydroxydopamine (6-OHDA) rat model of PD. AAV2-hIL-10 reduced the 6-OHDA-induced loss of tyrosine hydroxylase (TH)-positive neurons in the SNc, and also reduced loss of striatal dopamine (DA). Pretreatment with AAV2-hIL-10 reduced glial activation in the SNc but did not attenuate striatal release of the inflammatory cytokine IL-1beta. Assessment of rotational behavior in response to apomorphine challenge showed absence of asymmetry, confirming protection of dopaminergic innervation of the lesioned striatum. At baseline, 6-OHDA-lesioned animals displayed a deficit in contralateral forelimb use, but pretreatment with AAV2-hIL-10 reduced this forelimb akinesia. Transcriptional analyses revealed alteration of a few genes by AAV2-hIL-10; these alterations may contribute to neuroprotection. This study supports the need for further investigations relating to gene therapies aimed at reducing neuroinflammation in early PD.


Subject(s)
Disease Models, Animal , Genetic Therapy/methods , Interleukin-10/physiology , Parkinson Disease/therapy , Animals , Apomorphine/pharmacology , Behavior, Animal/drug effects , Chromatography, High Pressure Liquid , Dependovirus/genetics , Enzyme-Linked Immunosorbent Assay , Forelimb/drug effects , Forelimb/metabolism , Forelimb/physiopathology , Gene Expression/drug effects , Gene Expression Profiling , Genetic Vectors/genetics , Humans , Hydroxydopamines/pharmacology , Immunohistochemistry , Interleukin-10/genetics , Interleukin-10/metabolism , Male , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology
13.
Free Radic Biol Med ; 44(11): 1960-9, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18395527

ABSTRACT

Mitochondrial alterations have been associated with the cytotoxic effect of 6-hydroxydopamine (6-OHDA), a widely used neurotoxin to study Parkinson's disease. Herein we studied the potential effects of 6-OHDA on mitochondrial morphology in SH-SY5Y neuroblastoma cells. By immunofluorescence and time-lapse fluorescence microscopy we demonstrated that 6-OHDA induced profound mitochondrial fragmentation in SH-SY5Y cells, an event that was similar to mitochondrial fission induced by overexpression of Fis1p, a membrane adaptor for the dynamin-related protein 1 (DLP1/Drp1). 6-OHDA failed to induce any changes in peroxisome morphology. Biochemical experiments revealed that 6-OHDA-induced mitochondrial fragmentation is an early event preceding the collapse of the mitochondrial membrane potential and cytochrome c release in SH-SY5Y cells. Silencing of DLP1/Drp1, which is involved in mitochondrial and peroxisomal fission, prevented 6-OHDA-induced fragmentation of mitochondria. Furthermore, in cells silenced for Drp1, 6-OHDA-induced cell death was reduced, indicating that a block in mitochondrial fission protects SH-SY5Y cells against 6-OHDA toxicity. Experiments in mouse embryonic fibroblasts deficient in Bax or p53 revealed that both proteins are not essential for 6-OHDA-induced mitochondrial fragmentation. Our data demonstrate for the first time an involvement of mitochondrial fragmentation and Drp1 function in 6-OHDA-induced apoptosis.


Subject(s)
Apoptosis/drug effects , GTP Phosphohydrolases/metabolism , Hydroxydopamines/pharmacology , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Neuroblastoma/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Cytochromes c/metabolism , Dynamins , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fluorescent Antibody Technique , Humans , Membrane Potential, Mitochondrial , Mice , Mice, Knockout , Neuroblastoma/pathology , Tumor Suppressor Protein p53/physiology , bcl-2-Associated X Protein/physiology
14.
Cereb Cortex ; 17(1): 18-27, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16481566

ABSTRACT

We have previously demonstrated that prefrontal serotonin depletion impairs orbitofrontal cortex (OFC)-mediated serial discrimination reversal (SDR) learning but not lateral prefrontal cortex (PFC)-mediated attentional set shifting. To address the neurochemical specificity of this reversal deficit, Experiment 1 compared the effects of selective serotonin and selective dopamine depletions of the OFC on performance of the SDR task. Whereas serotonin depletions markedly impaired performance, OFC dopamine depletions were without effect. The behavioral specificity of this reversal impairment was investigated in Experiment 2 by examining the effect of OFC serotonin depletion on performance of a modified SDR task designed to distinguish between 3 possible causes of the impairment. The results showed that the reversal deficit induced by prefrontal serotonin depletion was not due to a failure to approach a previously unrewarded stimulus (enhanced learned avoidance) or reduced proactive interference. Instead, it was due specifically to a failure to inhibit responding to the previously rewarded stimulus. The neurochemical and behavioral specificity of this particular form of cognitive inflexibility is of particular relevance to our understanding of the aetiology and treatment of inflexible behavior apparent in many neuropsychiatric and neurodegenerative disorders involving the PFC.


Subject(s)
Behavior, Animal/physiology , Cognition/physiology , Prefrontal Cortex/physiology , Reversal Learning/physiology , Serotonin/physiology , 5,7-Dihydroxytryptamine/pharmacology , Animals , Avoidance Learning/physiology , Brain Chemistry/physiology , Callithrix , Color , Discrimination Learning/physiology , Dopamine/physiology , Female , Hydroxydopamines/pharmacology , Hydroxyindoleacetic Acid/metabolism , Male , Norepinephrine/metabolism , Photic Stimulation , Prefrontal Cortex/metabolism , Psychomotor Performance/physiology , Reward , Serial Learning/physiology , Serotonin Agents/pharmacology
15.
Stem Cells ; 25(4): 918-28, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17170065

ABSTRACT

The derivation of dopamine neurons is one of the best examples of the clinical potential of embryonic stem (ES) cells, but the long-term function of the grafted neurons has not been established. Here, we show that, after transplantation into an animal model, neurons derived from mouse ES cells survived for over 32 weeks, maintained midbrain markers, and had sustained behavioral effects. Microdialysis in grafted animals showed that dopamine (DA) release was induced by depolarization and pharmacological stimulants. Positron emission tomography measured the expression of presynaptic dopamine transporters in the graft and also showed that the number of postsynaptic DA D(2) receptors was normalized in the host striatum. These data suggest that ES cell-derived neurons show DA release and reuptake and stimulate appropriate postsynaptic responses for long periods after implantation. This work supports continued interest in ES cells as a source of functional DA neurons.


Subject(s)
Dopamine/physiology , Embryonic Stem Cells/physiology , Neurons/cytology , Neurons/physiology , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Stem Cell Transplantation , Animals , Cell Differentiation , Disease Models, Animal , Embryonic Stem Cells/pathology , Female , Hydroxydopamines/pharmacology , Immunohistochemistry , Mice , Microdialysis , Rats , Rats, Sprague-Dawley , Stereotyped Behavior
16.
Biochem Pharmacol ; 70(11): 1642-52, 2005 Nov 25.
Article in English | MEDLINE | ID: mdl-16226724

ABSTRACT

Antioxidants and iron chelating molecules are known as neuroprotective agents in animal models of neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD). In this study, we designed and synthesized a novel bifunctional molecule (M10) with radical scavenging and iron chelating ability on an amino acid carrier likely to be a substrate for system L, thus targeting the compound to the central nervous system (CNS). M10 had a moderate iron affinity in HEPES buffer (pH 7.4) with logK(3)=12.25+/-0.55 but exhibited highly inhibitory action against iron-induced lipid peroxidation, with an IC(50) value (12microM) comparable to that of desferal (DFO). EPR studies indicated that M10 was a highly potent *OH scavenger with an IC(50) of about 0.3 molar ratio of M10 to H(2)O(2). In PC12 cell culture, M10 was at least as potent as the anti-Parkinson drug rasagiline in protecting against cell death induced by serum-deprivation and by 6-hydroxydopamine (6-OHDA). These results suggest that M10 deserves further investigation as a potential agent for the treatment of neurodegenerative disorders such as AD and PD.


Subject(s)
Amino Acids/metabolism , Iron Chelating Agents/pharmacology , Iron/metabolism , Neurons/drug effects , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Alanine/analogs & derivatives , Alanine/chemistry , Alanine/pharmacology , Animals , Apoptosis/drug effects , Culture Media, Serum-Free/pharmacology , Drug Evaluation, Preclinical , Free Radical Scavengers/pharmacology , Hydroxydopamines/pharmacology , Hydroxyl Radical/metabolism , Hydroxyquinolines/chemistry , Hydroxyquinolines/pharmacology , Iron Chelating Agents/metabolism , Lipid Peroxidation/drug effects , Molecular Structure , Neurons/metabolism , PC12 Cells , Rats
17.
Eur J Neurosci ; 22(5): 1158-68, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16176358

ABSTRACT

Vascular inflammation is well known for its ability to compromise the function of the blood--brain barrier (BBB). Whether inflammation on the parenchymal side of the barrier, such as that associated with Parkinson's-like dopamine (DA) neuron lesions, similarly disrupts BBB function, is unknown. We assessed BBB integrity by examining the leakage of FITC-labeled albumin or horseradish peroxidase from the vasculature into parenchyma in animals exposed to the DA neurotoxin 6-hydroxydopamine (6OHDA). Unilateral injections of 6OHDA into the striatum or the medial forebrain bundle produced increased leakage in the ipsilateral substantia nigra and striatum 10 and 34 days following 6OHDA. Microglia were markedly activated and DA neurons were reduced by the lesions. The areas of BBB leakage were associated with increased expression of P-glycoprotein and beta 3-integrin expression suggesting, respectively, a compensatory response to inflammation and possible angiogenesis. Behavioural studies revealed that domperidone, a DA antagonist that normally does not cross the BBB, attenuated apomorphine-induced stereotypic behaviour in animals with 6OHDA lesions. This suggests that drugs which normally have no effect in brain can enter following Parkinson-like lesions. These data suggest that the events associated with DA neuron loss compromise BBB function.


Subject(s)
Blood-Brain Barrier/drug effects , Hydroxydopamines/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Albumins/metabolism , Animals , Blood-Brain Barrier/physiology , Cell Count/methods , Dopamine/metabolism , Functional Laterality , Horseradish Peroxidase/metabolism , Immunohistochemistry/methods , Integrin beta3/metabolism , Male , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Time Factors , Tyrosine 3-Monooxygenase/metabolism
18.
Gene ; 331: 149-57, 2004 Apr 28.
Article in English | MEDLINE | ID: mdl-15094201

ABSTRACT

Dopamine denervation in the striata of patients with Parkinson's disease (PD) leads to changes in neural plasticity. However, the mechanisms leading to the changes are still poorly understood. In an effort to study the molecular events in the denervated striatum, we identified and cloned rat karyopherin alpha 1 (KPNA1), a member of the importin/karyopherin alpha (KPNA) family. DNA sequence analysis revealed that the full-length cDNA, encoding rat KPNA1, was 4975 bp with a short 5'-untranslated region (UTR) of 70 bp, a putative coding sequence of 1617 bp, and an unusually long 3'-UTR of 3266 bp. The gene shared a high degree of similarity with its mouse and human homologs at both cDNA and protein levels. By computational analysis of its genomic sequence, the transcription unit was shown to span a 44-kb region and consist of 13 exons varying in size from 89 (6th exon) to 3454 bp (13th exon), and 12 introns varying in size from 0.3 to 8.9 kb. Reverse transcriptase-polymerase chain reaction (RT-PCR) analysis demonstrated that KPNA1 transcript existed in various adult tissues. Both Northern blot and semi-quantitative RT-PCR analysis showed that the expression level of KPNA1 mRNA was altered in the denervated striatum post-lesion in a time-dependent manner, reaching the maximum at 2 weeks post-lesion. Our results suggest involvement of KPNA1 in the striatal responses to denervation following 6-hydroxydopamine (6-OHDA)-induced lesion.


Subject(s)
Gene Expression Profiling , alpha Karyopherins/genetics , 3' Flanking Region/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Corpus Striatum/drug effects , Corpus Striatum/metabolism , DNA, Complementary/chemistry , DNA, Complementary/genetics , Databases, Nucleic Acid , Exons , Female , Gene Expression/drug effects , Genes/genetics , Hydroxydopamines/pharmacology , Introns , Molecular Sequence Data , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid
19.
Ann N Y Acad Sci ; 973: 70-4, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12485836

ABSTRACT

We show that the glial cell line-derived neurotrophic factor (GDNF) activates the PI3K/Akt-signaling pathway in human neuroblastoma cells that express functional Ret-receptor complexes. Consistent with this finding we show PI3K-dependent Bad-inactivation by binding to 14-3-3 proteins in response to GDNF. Using differential display techniques we detected several cDNA clones differentially expressed after treatment with GDNF or 6-OHDA.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Nerve Growth Factors/pharmacology , Neuroprotective Agents/pharmacology , Signal Transduction/physiology , 14-3-3 Proteins , Glial Cell Line-Derived Neurotrophic Factor , Humans , Hydroxydopamines/pharmacology , Neoplasm Proteins/genetics , Neuroblastoma , Phosphatidylinositol 3-Kinases/metabolism , Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Cells, Cultured , Tyrosine 3-Monooxygenase/metabolism
20.
Redox Rep ; 7(3): 171-7, 2002.
Article in English | MEDLINE | ID: mdl-12189048

ABSTRACT

Green tea polyphenols exert a wide range of biochemical and pharmacological effects, and have been shown to possess antimutagenic and anticarcinogenic properties. Oxidative stress is involved in the pathogenesis of Parkinson's disease. However, although green tea polyphenols may be expected to inhibit the progression of Parkinson's disease on the basis of their known antioxidant activity, this has not previously been established. In the present study, we evaluated the neuroprotective effects of green tea polyphenols in the Parkinson's disease pathological cell model. The results show that the natural antioxidants have significant inhibitory effects against apoptosis induced by oxidative stress. 6-Hydroxydopamine (6-OHDA)-induced apoptosis in catecholaminergic PC12 cells was chosen as the in vitro model of Parkinson's disease in our study. Apoptotic characteristics of PC12 cells were assessed by MTT assay, flow cytometry, fluorescence microscopy and DNA fragmentation. Green tea polyphenols and their major component, EGCG at a concentration of 200 microM, exert significant protective effects against 6-OHDA-induced PC12 cell apoptosis. EGCG is more effective than the mixture of green tea polyphenols. The antioxidant function of green tea polyphenols may account for this neuroprotective effect. The present study supports the notion that green tea polyphenols have the potential to be effective as neuropreventive agents for the treatment of neurodegenerative diseases.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Catechin/analogs & derivatives , Catechin/pharmacology , Flavonoids , Hydroxydopamines/pharmacology , Plant Extracts/pharmacology , Tea , Animals , Cell Survival/drug effects , Kinetics , PC12 Cells , Phenols/pharmacology , Pheochromocytoma , Polymers/pharmacology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...