Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Brain Behav Immun ; 69: 456-469, 2018 03.
Article in English | MEDLINE | ID: mdl-29339319

ABSTRACT

The impact of treatment with the noradrenaline (NA) re-uptake inhibitor atomoxetine and the α2-adrenoceptor (AR) antagonist idazoxan in an animal model of Parkinson's disease (PD) was assessed. Concurrent systemic treatment with atomoxetine and idazoxan, a combination which serves to enhance the extra-synaptic availability of NA, exerts anti-inflammatory and neuroprotective effects following delivery of an inflammatory stimulus, the bacterial endotoxin, lipopolysaccharide (LPS) into the substantia nigra. Lesion-induced deficits in motor function (akinesia, forelimb-use asymmetry) and striatal dopamine (DA) loss were rescued to varying degrees depending on the treatment. Treatment with atomoxetine following LPS-induced lesion to the substantia nigra, yielded a robust anti-inflammatory effect, suppressing microglial activation and expression of the pro-inflammatory cytokine TNF-α whilst increasing the expression of neurotrophic factors. Furthermore atomoxetine treatment prevented loss of tyrosine hydroxylase (TH) positive nigral dopaminergic neurons and resulted in functional improvements in motor behaviours. Atomoxetine alone was sufficient to achieve most of the observed effects. In combination with idazoxan, an additional improvement in the impairment of contralateral limb use 7 days post lesion and a reduction in amphetamine-mediated rotational asymmetry 14 days post-lesion was observed, compared to atomoxetine or idazoxan treatments alone. The results indicate that increases in central NA tone has the propensity to regulate the neuroinflammatory phenotype in vivo and may act as an endogenous neuroprotective mechanism where inflammation contributes to the progression of DA loss. In accordance with this, the clinical use of agents such as NA re-uptake inhibitors and α2-AR antagonists may prove useful in enhancing the endogenous neuroimmunomodulatory potential of NA in conditions associated with brain inflammation.


Subject(s)
Atomoxetine Hydrochloride/pharmacology , Brain/drug effects , Dopaminergic Neurons/drug effects , Idazoxan/pharmacology , Motor Activity/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/drug therapy , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic Uptake Inhibitors/therapeutic use , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Animals , Atomoxetine Hydrochloride/therapeutic use , Brain/metabolism , Brain/pathology , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Idazoxan/therapeutic use , Lipopolysaccharides , Neuroprotective Agents/therapeutic use , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Rats , Rats, Wistar , Treatment Outcome
2.
Pain ; 158(1): 149-160, 2017 01.
Article in English | MEDLINE | ID: mdl-27984527

ABSTRACT

Antidepressants are first-line treatments of neuropathic pain but not all these drugs are really effective. Agomelatine is an antidepressant with a novel mode of action, acting as an MT1/MT2 melatonergic receptor agonist and a 5-HT2C receptor antagonist that involves indirect norepinephrine release. Melatonin, serotonin, and norepinephrine have been involved in the pathophysiology of neuropathic pain. Yet, no study has been conducted to determine agomelatine effects on neuropathic pain in animal models. Using 3 rat models of neuropathic pain of toxic (oxaliplatin/OXA), metabolic (streptozocin/STZ), and traumatic (sciatic nerve ligation/CCI [chronic constriction nerve injury]) etiologies, we investigated the antihypersensitivity effect of acute and repeated agomelatine administration. We then determined the influence of melatonergic, 5-HT2C, α-2 and ß-1/2 adrenergic receptor antagonists in the antihypersensitivity effect of agomelatine. The effect of the combination of agomelatine + gabapentin was evaluated using an isobolographic approach. In STZ and CCI models, single doses of agomelatine significantly and dose dependently reduced mechanical hypersensitivity. After daily administrations for 2 weeks, this effect was confirmed in the CCI model and agomelatine also displayed a marked antihypersensitivity effect in the OXA model. The antihypersensitivity effect of agomelatine involved melatonergic, 5-HT2C, and α-2 adrenergic receptors but not beta adrenoceptors. The isobolographic analysis demonstrated that the combination of agomelatine + gabapentin had additive effects. Agomelatine exerts a clear-cut antihypersensitivity effect in 3 different neuropathic pain models. Its effect is mediated by melatonergic and 5-HT2C receptors and, although agomelatine has no affinity, also by α-2 adrenergic receptors. Finally, agomelatine combined with gabapentin produces an additive antihypersensitivity effect.


Subject(s)
Acetamides/therapeutic use , Hypnotics and Sedatives/therapeutic use , Neuralgia/drug therapy , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Amines/therapeutic use , Animals , Antineoplastic Agents/toxicity , Constriction, Pathologic/complications , Cyclohexanecarboxylic Acids/therapeutic use , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Excitatory Amino Acid Antagonists/therapeutic use , Gabapentin , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Idazoxan/therapeutic use , Male , Motor Activity/drug effects , Motor Activity/physiology , Neuralgia/etiology , Organoplatinum Compounds/toxicity , Oxaliplatin , Pain Measurement , Rats , Rats, Sprague-Dawley , Thiophenes/therapeutic use , gamma-Aminobutyric Acid/therapeutic use
3.
Eur J Pharmacol ; 736: 70-6, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-24797785

ABSTRACT

We have previously shown that Idazoxan (IDA), an imidazoline 2 receptor ligand, is neuroprotective against spinal cord injury caused by experimental autoimmune encephalomyelitis (EAE) in mouse, an animal modal of multiple sclerosis (MS). However, the protective mechanism remains unclear. Here, we provided evidence to show that IDA confers neuroprotection through reduction in blood-brain barrier (BBB) damage. EAE was induced by immunizing C57 BL/6 mice with myelin oligodendrocyte glycoprotein35-55 amino acid peptide (MOG35-55). IDA was administrated for 14 days after MOG immunization at 2 mg/kg (i.p., bid). Significant reduction in BBB damage occurred in the IDA-treated group of mice compared with the saline-treated group, as evidenced by the reduction in Evan׳s blue content in the brain tissue and the reduced BBB tight junction damage viewed under a transmission electron microscope. Moreover, EAE-induced reductions in tight junction proteins (JAM-1, Occludin, Claudin-5 and ZO-1) were also significantly ameliorated in IDA-treated mice, all of which supported the notion that IDA reduced BBB damage. Interestingly, the expression levels of extracellular matrix metalloproteinase-9 (MMP-9) and the ratio of MMP-9 against tissue inhibitor of metalloproteinase-1 (TIMP-1), which is known to be associated with MS-induced BBB damage, were significantly reduced in IDA-treated group, lending further support to the hypothesis that IDA confers brain protection through reducing BBB damage. This study raised a possibility that IDA is a promising pro-drug for development against MS.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Blood-Brain Barrier/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Idazoxan/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Behavior, Animal/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/ultrastructure , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Idazoxan/pharmacology , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Neuroprotective Agents/pharmacology , Permeability/drug effects , Tight Junctions/drug effects , Tight Junctions/pathology , Tissue Inhibitor of Metalloproteinase-1/metabolism
4.
Pharmacol Biochem Behav ; 100(3): 607-15, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21978941

ABSTRACT

While L-3,4-dihydroxyphenylalanine (L-DOPA) remains the standard treatment for Parkinson's disease (PD), long-term efficacy is often compromised by L-DOPA-induced dyskinesia (LID). Recent research suggests that targeting the noradrenergic (NE) system may provide relief from both PD and LID, however, most PD patients exhibit NE loss which may modify response to such strategies. Therefore this investigation aimed to characterize the development and expression of LID and the anti-dyskinetic potential of the α2- and ß-adrenergic receptor antagonists idazoxan and propranolol, respectively, in rats receiving 6-OHDA lesions with (DA lesion) or without desipramaine protection (DA+NE lesion). Male Sprague-Dawley rats (N=110) received unilateral 6-hydroxydopamine lesions. Fifty-three rats received desipramine to protect NE neurons (DA lesion) and 57 received no desipramine reducing striatal and hippocampal NE content 64% and 86% respectively. In experiment 1, the development and expression of L-DOPA-induced abnormal involuntary movements (AIMs) and rotations were examined. L-DOPA efficacy using the forepaw adjusting steps (FAS) test was also assessed in DA- and DA+NE-lesioned rats. In experiment 2, DA- and DA+NE-lesioned rats received pre-treatments of idazoxan or propranolol followed by L-DOPA after which the effects of these adrenergic compounds were observed. Results demonstrated that moderate NE loss reduced the development and expression of AIMs and rotations but not L-DOPA efficacy while anti-dyskinetic efficacy of α2- and ß-adrenergic receptor blockade was maintained. These findings suggest that the NE system modulates LID and support the continued investigation of adrenergic compounds for the improved treatment of PD.


Subject(s)
Adrenergic Neurons/drug effects , Adrenergic alpha-Antagonists/therapeutic use , Adrenergic beta-Antagonists/therapeutic use , Corpus Striatum/physiopathology , Dyskinesia, Drug-Induced/drug therapy , Hippocampus/physiopathology , Levodopa/adverse effects , Animals , Behavior, Animal/drug effects , Corpus Striatum/drug effects , Desipramine/pharmacology , Disease Models, Animal , Dyskinesia, Drug-Induced/physiopathology , Hippocampus/drug effects , Idazoxan/therapeutic use , Levodopa/therapeutic use , Male , Molecular Targeted Therapy , Neuroprotective Agents/pharmacology , Oxidopamine , Parkinson Disease/drug therapy , Parkinson Disease/physiopathology , Propranolol/therapeutic use , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Sympathectomy, Chemical
5.
Pharmacology ; 88(5-6): 233-41, 2011.
Article in English | MEDLINE | ID: mdl-21997570

ABSTRACT

BACKGROUND: Centhaquin is a centrally acting hypotensive drug like clonidine. Clonidine also produces analgesia and hypothermia in mice and potentiates morphine analgesia. Clonidine analgesia is blocked by idazoxan and naloxone while it is potentiated by BQ123 and sulfisoxazole. This study was conducted to determine the analgesic and hypothermic properties of centhaquin, and to assess whether it potentiates morphine analgesia. Yohimbine (α(2)-adrenergic antagonist), idazoxan (imidazoline/α(2)-adrenergic antagonist), naloxone (opioid antagonist), and BQ123 and sulfisoxazole (endothelin ET(A) antagonists) were used to study the involvement of these receptors in centhaquin analgesia and hypothermia. METHODS: Analgesic (tail flick and hot-plate tests) latencies and body temperatures were measured in male Swiss Webster mice treated with vehicle plus centhaquin, antagonists plus centhaquin or centhaquin plus morphine. RESULTS: Centhaquin produced dose-dependent analgesia which was partially blocked by yohimbine, idazoxan and naloxone. BQ123 and sulfisoxazole did not affect centhaquin analgesia. Morphine analgesia was not potentiated by centhaquin. Centhaquin produced mild hypothermia which was not blocked by yohimbine, idazoxan, naloxone, BQ123 or sulfisoxazole. CONCLUSIONS: This is the first report demonstrating the analgesic activity of centhaquin. The α(2)-adrenergic, imidazoline and opioid receptors are involved in mediating centhaquin analgesia. Endothelin ET(A) receptors do not play a role in centhaquin analgesia; centhaquin does not augment morphine analgesia.


Subject(s)
Analgesics/therapeutic use , Imidazoline Receptors/metabolism , Pain/drug therapy , Piperazines/therapeutic use , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Opioid/metabolism , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Analgesics, Opioid/therapeutic use , Animals , Antihypertensive Agents/therapeutic use , Body Temperature/drug effects , Drug Interactions , Endothelin A Receptor Antagonists , Hot Temperature , Idazoxan/therapeutic use , Imidazoline Receptors/antagonists & inhibitors , Male , Mice , Morphine/therapeutic use , Naloxone/therapeutic use , Narcotic Antagonists , Receptor, Endothelin A/metabolism , Yohimbine/therapeutic use
6.
J Neurochem ; 112(2): 444-52, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19895663

ABSTRACT

L-DOPA-induced dyskinesia is characterised by debilitating involuntary movement, which limits quality of life in patients suffering from Parkinson's disease. Here, we investigate effects of the a2 adrenoceptor antagonist idazoxan on L-DOPA-induced dyskinesia as well as on alterations of extracellular L-DOPA and dopamine (DA) levels in the striatum in dyskinetic rats. Male Wistar rats were unilaterally lesioned with 6-hydroxydopamine and subsequently treated with L-DOPA/benserazide to induce stable dyskinetic movements.Administration of idazoxan [(9 mg/kg, intraperitoneal (i.p.)]significantly alleviated L-DOPA-induced dyskinesia, whereas idazoxan (3 mg/kg, i.p.) did not affect dyskinetic behaviour.Bilateral in vivo microdialysis revealed that idazoxan 9 mg/kg reduces extracellular peak L-DOPA levels in the lesioned and intact striatum as well as DA levels in the lesioned striatum. In parallel, the exposure to idazoxan in the striatum was monitored.Furthermore, no idazoxan and L-DOPA drug-drug interaction was found in plasma, brain tissue and CSF. In conclusion, the decrease of L-DOPA-derived extracellular DA levels in the lesioned striatum significantly contributes to the anti-dyskinetic effect of idazoxan.


Subject(s)
Adrenergic alpha-Antagonists/therapeutic use , Corpus Striatum/metabolism , Dopamine/metabolism , Dyskinesia, Drug-Induced/drug therapy , Idazoxan/therapeutic use , Adrenergic Agents/toxicity , Adrenergic alpha-Antagonists/pharmacokinetics , Analysis of Variance , Animals , Antiparkinson Agents/adverse effects , Antiparkinson Agents/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Corpus Striatum/drug effects , Disease Models, Animal , Dyskinesia, Drug-Induced/etiology , Idazoxan/pharmacokinetics , Levodopa/adverse effects , Levodopa/pharmacokinetics , Male , Microdialysis/methods , Neurotoxicity Syndromes/complications , Neurotoxicity Syndromes/drug therapy , Neurotoxicity Syndromes/etiology , Oxidopamine/toxicity , Rats , Rats, Wistar , Tandem Mass Spectrometry/methods
7.
Neurol Res ; 31(4): 390-5, 2009 May.
Article in English | MEDLINE | ID: mdl-19508825

ABSTRACT

OBJECTIVE: The aim of this study was to determine whether 2-(2-benzofuranyl)-2-imidazoline, an imidazoline I(2) receptor ligand, could protect against cell death from brain injury and improve the functional outcome after focal cerebral ischemia in rats. METHODS: Transient focal ischemia was induced by suture occlusion of the middle cerebral artery. Rats were intraperitoneally treated with a vehicle, 2-(2-benzofuranyl)-2-imidazoline or idazoxan immediately after focal ischemia. Infarct volume was assessed by 2,3,5-triphenyltrazolium chloride staining and neurobehavioral deficits were monitored. The volume of cell death in the penumbra after ischemia was determined by immunostaining using anti-cleaved caspase-3 antibody and terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling (TUNEL). RESULTS: Both 2-(2-benzofuranyl)-2-imidazoline and idazoxan significantly improved the neurological score compared with the vehicle at 24 hours after focal ischemia. Treatment with 2-(2-benzofuranyl)-2-imidazoline or idazoxan also significantly reduced infarct volume and the number of both caspase-3- and TUNEL-positive cells in the penumbra compared with vehicle-treated rats (p<0.01 and p<0.05, respectively). CONCLUSION: The results suggest the neuroprotective role of 2-(2-benzofuranyl)-2-imidazoline and idazoxan in focal cerebral ischemia, and may therefore represent useful targets for developing new treatments for stroke.


Subject(s)
Benzofurans/therapeutic use , Brain Injuries/prevention & control , Brain Ischemia/drug therapy , Imidazoles/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Brain Ischemia/complications , Caspase 3/metabolism , Disease Models, Animal , Idazoxan/therapeutic use , Imidazoline Receptors , In Situ Nick-End Labeling/methods , Male , Neurologic Examination/methods , Rats , Rats, Sprague-Dawley , Tetrazolium Salts
8.
Clin Exp Immunol ; 156(2): 353-62, 2009 May.
Article in English | MEDLINE | ID: mdl-19250273

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by heavy production of proinflammatory cytokines such as tumour necrosis factor (TNF)-alpha and interleukin (IL)-1beta. Interactions of the autonomic nervous system with local immune cells play an important role in the development of IBD, and the balance of autonomic nerve function is broken in IBD patients with sympathetic overactivity. However, the function of catecholamines in the progress of colitis is unclear. In this study, we examined the role of catecholamines via alpha2-adrenoreceptor in acute murine colitis. The expression of tyrosine hydroxylase (TH) and dopamine b-hydroxylase (DBH), two rate-limiting enzymes in catecholamine synthesis, was detected by immunohistochemistry in murine colitis. Murine colitis was induced by dextran sodium sulphate or trinitrobenzene sulphonic acid (TNBS), and the mice were administered RX821002 or UK14304, alpha2-adrenoceptor antagonists or agonists. Colitis was evaluated by clinical symptoms, myeloperoxidase assay, TNF-alpha and IL-1beta production and histology. Lamina propria mononuclear cells (LPMCs) from mice with TNBS colitis were cultured in the absence or presence of RX821002 or UK14304, and stimulated further by lipopolysaccharide. TH and DBH are induced in LPMCs of inflamed colon, the evidence of catecholamine synthesis during the process of colitis. RX821002 down-regulates the production of proinflammatory cytokines from LPMCs, while UK14304 leads to exacerbation of colitis. Together, our data show a critical role of catecholamines via alpha2-adrenoreceptors in the progress of acute colitis, and suggest that use of the alpha2-adrenoceptor antagonist represents a novel therapeutic approach for the management of colitis.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists , Colitis/drug therapy , Colon , Idazoxan/analogs & derivatives , Intestinal Mucosa/immunology , Adrenergic alpha-Agonists/pharmacology , Animals , Brimonidine Tartrate , Cells, Cultured , Colitis/enzymology , Colitis/immunology , Cytokines/immunology , Dextran Sulfate , Dopamine beta-Hydroxylase/metabolism , Idazoxan/therapeutic use , Immunohistochemistry , Intestinal Mucosa/enzymology , Male , Mice , Mice, Inbred BALB C , Models, Animal , Quinoxalines/pharmacology , Tyrosine 3-Monooxygenase/metabolism
9.
Brain Res ; 1253: 198-209, 2009 Feb 09.
Article in English | MEDLINE | ID: mdl-19083996

ABSTRACT

Idazoxan, an imidazoline 2 receptor (I(2)R) ligand, has been shown to protect against brain injury in several animal models of neurological disorders. In the present study we investigated the effect of idazoxan on experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. EAE was induced by immunizing Wistar rats with guinea pig spinal cord homogenates emulsified in CFA, followed by daily treatment of idazoxan (0, 0.5 mg/kg, 1.5 mg/kg, 4.5 mg/kg, i.p, bid) for 10 days. The results showed that the treatment of idazoxan (1.5 mg/kg and 4.5 mg/kg) significantly decreased the incidence and alleviated inflammatory cell infiltration and demyelination in spinal cords and cerebral cortex. Furthermore, the protective effect of idazoxan on EAE was associated with the enhanced astrocytic activation and attenuated microglial activation and with the subsequent down-regulated expression of proinflammatory cytokines IL-12p40 and IFN-gamma and up-regulated expression of anti-inflammatory cytokines IL-10 and TGF-beta(1). Thus, the daily treatment of the I(2)R ligand idazoxan for 10 days attenuates EAE pathology by differential modulation of astrocytic and microglial activations, raising a possibility that the I(2)R ligand may be a novel strategy for treating EAE.


Subject(s)
Astrocytes/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Idazoxan/therapeutic use , Microglia/drug effects , Spinal Cord/physiopathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Astrocytes/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cytokines/genetics , Cytokines/metabolism , Demyelinating Diseases/drug therapy , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Gene Expression Regulation , Imidazoline Receptors/agonists , Imidazoline Receptors/metabolism , Microglia/physiology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology , Multiple Sclerosis/physiopathology , Neuroprotective Agents/therapeutic use , RNA, Messenger/metabolism , Rats , Rats, Wistar , Spinal Cord/pathology
10.
Int J Neuropsychopharmacol ; 8(3): 315-27, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15857571

ABSTRACT

The alpha(2) adrenoceptor antagonist idazoxan enhances antipsychotic efficacy of classical dopamine D(2) antagonists in treatment-resistant schizophrenia. The mechanisms are not fully understood, but we have previously shown that the combination of idazoxan with the D(2/3) receptor antagonist raclopride, similarly to clozapine but not classical antipsychotic drugs, augments dopamine efflux in the prefrontal cortex, and also generates an enhanced suppression of the conditioned avoidance response. We have now investigated the effects of clozapine, raclopride, idazoxan and the combination of raclopride and idazoxan on (i) electrically evoked excitatory post-synaptic potentials and currents in pyramidal cells of the rat medial prefrontal cortex, using intracellular electrophysiological recording in vitro, (ii) the impaired cognitive function induced by the selective N-methyl-D-aspartate (NMDA) receptor antagonist MK-801, using the 8-arm radial maze test, (iii) the in-vivo D2, alpha(2A) and alpha(2C) receptor occupancies of these pharmacological treatments, using ex-vivo autoradiography. Whereas neither idazoxan nor raclopride alone had any effect, the combination exerted the same facilitation of glutamatergic transmission in rat prefrontal pyramidal neurons as clozapine, and this effect was found to be mediated by dopamine acting at D(1) receptors. Similarly to clozapine, the combination of idazoxan and raclopride also completely reversed the working-memory impairment in rats induced by MK-801. Moreover, these effects of the two treatment regimes were obtained at similar occupancies at D(2), alpha(2A) and alpha(2C) receptors respectively. Our results provide novel neurobiological and behavioural support for a pro-cognitive effect of adjunctive use of idazoxan with antipsychotic drugs that lack appreciable alpha(2) adrenoceptor-blocking properties, and define presynaptic alpha(2) adrenoceptors as major targets in antipsychotic drug development.


Subject(s)
Adrenergic alpha-Antagonists/therapeutic use , Cerebral Cortex/metabolism , Cognition Disorders/drug therapy , Dopamine D2 Receptor Antagonists , Glutamic Acid/metabolism , Synaptic Transmission/physiology , Animals , Behavior, Animal , Cerebral Cortex/drug effects , Clozapine/therapeutic use , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Idazoxan/therapeutic use , In Vitro Techniques , Male , Maze Learning/drug effects , Raclopride/therapeutic use , Radioligand Assay/methods , Rats , Rats, Sprague-Dawley , Rats, Wistar , Synaptic Transmission/drug effects
11.
Exp Neurol ; 191(2): 243-50, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15649479

ABSTRACT

l-DOPA-induced dyskinesia (LID) remains a major complication of the treatment of Parkinson's disease (PD). Whilst the MPTP-lesioned primate provides an excellent animal model in which to develop new therapies, however, it is logistically difficult to employ widely. Thus, a simple rodent assay to screen multiple compounds as candidates for further study of their potential in LID would be a valuable addition to the drug development process. Here, we investigate how agents with demonstrated ability to reduce LID in man and monkey can regulate l-DOPA-induced behaviours in the reserpine-treated rat. Administration of l-DOPA (125 mg/kg) to reserpine-treated rats elicited high levels of both horizontal and vertical movement. Drugs that have previously been found to reduce LID in parkinsonian primates and PD patients without compromising the anti-parkinsonian efficacy of l-DOPA selectively and dose-dependently reduce vertical components of activity when co-administered with l-DOPA in the reserpine-treated rat. For instance, amantadine (1 mg/kg) and idazoxan (3 mg/kg) reduced vertical activity by 59% and 83%, respectively, while neither drug had significant effects on horizontal activity. In contrast, haloperidol (1 mg/kg), an agent lacking the ability to selectively reduce LID without compromising the anti-parkinsonian actions of l-DOPA, reduced both horizontal and vertical activity, by 98% and 99%, respectively. We also assessed the actions of an NMDA antagonist, a class of compound proposed to have potential as anti-dyskinetic agents. The effects of MK-801 were dose-dependent (0.01-0.5 mg/kg), at some doses (e.g., 0.05 mg/kg), providing selective reduction of vertical activity (90%), at others (e.g., 0.5 mg/kg), non-selective reduction of vertical and horizontal (99% and 77%, respectively). These observations highlight the association between potential anti-dyskinetic action and a selective reduction in l-DOPA-induced vertical activity in the reserpine-treated rat.


Subject(s)
Adrenergic alpha-Antagonists/therapeutic use , Anti-Dyskinesia Agents/therapeutic use , Dopamine Agents/therapeutic use , Drug Evaluation, Preclinical/methods , Dyskinesia, Drug-Induced/drug therapy , Levodopa/adverse effects , Adrenergic Uptake Inhibitors/pharmacology , Amantadine/therapeutic use , Animals , Antiparkinson Agents/adverse effects , Antiparkinson Agents/therapeutic use , Dizocilpine Maleate/therapeutic use , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/therapeutic use , Haloperidol/therapeutic use , Idazoxan/therapeutic use , Levodopa/therapeutic use , Male , Motor Activity/drug effects , Parkinson Disease/drug therapy , Rats , Rats, Sprague-Dawley , Reserpine/pharmacology
12.
Behav Brain Res ; 154(2): 353-63, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15313023

ABSTRACT

Noradrenaline, not only functions as a synaptic transmitter, but also promotes neural differentiation and regenerative processes. In Parkinson's disease, besides the dopaminergic degeneration, noradrenergic neurons of locus coeruleus origin degenerate as well. Drugs enhancing noradrenergic transmission in the locus coeruleus (e.g. alpha2-adrenoceptor antagonists) have been shown to be neuroprotective against Huntington's and ischemic animal models. However, in Parkinsonian animal models, most of the studies evaluated the worsening of experimental nigral neurodegeneration after locus coeruleus lesions. Here, it has been tested, whether treatment with the selective alpha2-adrenoceptor antagonist, 2-methoxy idazoxan (2.5 mg/kg i.p., twice daily for 5 days), before an experimental lesion to nigra, protects dopaminergic neurodegeneration. Dopaminergic degeneration was produced by 6-hydroxydopamine lesion in the median forebrain bundle. The concentrations of dopamine, 5-hydroxytryptamine and its metabolites were analysed in the various regions of the basal ganglia. The concentrations of noradrenaline and dopamine were measured in the regions innervated by locus coeruleus neurons and in the basal ganglia respectively, after 2-methoxy idazoxan treatment. The Parkinsonian behavior was assessed by catalepsy and activity test. 2-Methoxy idazoxan specifically increased the concentration of noradrenaline in the brain regions, innervated by locus coeruleus neurons. 6-OHDA lesion strongly depleted the concentration of dopamine and its metabolites in the striatum and SN, producing catalepsy and hypoactivity. Multiple treatments with 2-methoxy idazoxan reduced some of the observed neurochemical and behavioral indices of 6-hydroxydopamine-induced Parkinsonism, indicating neuroprotection. Although the mechanism underlying the neuroprotective property remains elusive, the therapeutic usage of alpha2-antagonists might be helpful in slowing the neuronal death and progression of Parkinson's disease.


Subject(s)
Adrenergic alpha-Antagonists/therapeutic use , Brain Chemistry/drug effects , Idazoxan/analogs & derivatives , Idazoxan/therapeutic use , Parkinsonian Disorders/prevention & control , Adrenergic alpha-Antagonists/pharmacology , Analysis of Variance , Animals , Catalepsy/drug therapy , Disease Models, Animal , Dopamine/metabolism , Drug Interactions , Hydroxyindoleacetic Acid/metabolism , Idazoxan/pharmacology , Male , Motor Activity/drug effects , Norepinephrine/metabolism , Oxidopamine , Parkinsonian Disorders/chemically induced , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Serotonin/metabolism , Statistics, Nonparametric
13.
Naunyn Schmiedebergs Arch Pharmacol ; 369(6): 629-38, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15118809

ABSTRACT

The alpha2-adrenoreceptor antagonist idazoxan counteracts catalepsy induced by neuroleptic agents and improves Parkinsonian signs in 1-methyl-4-phenyl 1,2,3,6,tetrahydropyridine-treated monkeys and in patients. The present study addressed the question of whether systemic administration of idazoxan (1.5 mg/kg i.p.) improves Parkinsonian symptoms in a rat model of permanent dopaminergic neurodegeneration. Dopaminergic degeneration was induced by injection of 6-hydroxydopamine (6-OHDA) into the medial forebrain bundle (MFB). Parkinsonian behaviour was assessed by catalepsy and open-field exploratory behaviour tests. Since dopaminergic and serotonergic mechanisms are thought to account for the anti-cataleptic/anti-Parkinsonian property of idazoxan, dopamine, 5-hydroxytryptamine (5-HT) and its metabolites in the regions of the basal ganglia and prefrontal cortex were analysed by HPLC. 6-OHDA lesions in the MFB produced catalepsy and hypoactivity in the open field and depleted dopamine and its metabolites in the basal ganglia and prefrontal cortex, but did not affect 5-HT. Treatment with idazoxan counteracted the observed Parkinsonian behaviour in 6-OHDA-lesioned rats and increased the spontaneous open-field activity in control rats. In both 6-OHDA and control animals, idazoxan increased DA level in the prefrontal cortex, but not in any other structures including the striatum. Idazoxan also increased the levels of 5-HT in the anterior striatum, prefrontal cortex and the ventral tegmental area of both 6-OHDA and control animals. These findings indicate that systemic administration of idazoxan counteracts 6-OHDA-induced Parkinsonian symptoms in rats and that both dopaminergic and serotonergic mechanisms could contribute to its anti-Parkinsonian effect.


Subject(s)
Adrenergic alpha-Antagonists/pharmacology , Antiparkinson Agents/pharmacology , Brain/drug effects , Idazoxan/pharmacology , Parkinsonian Disorders/prevention & control , Adrenergic alpha-Antagonists/therapeutic use , Animals , Brain/metabolism , Catalepsy/drug therapy , Disease Models, Animal , Dopamine/analysis , Dopamine/metabolism , Exploratory Behavior/drug effects , Hydroxyindoleacetic Acid/metabolism , Idazoxan/therapeutic use , Male , Motor Activity/drug effects , Oxidopamine , Parkinsonian Disorders/chemically induced , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Serotonin/analysis , Serotonin/metabolism , Time Factors
14.
Neuropsychopharmacology ; 28(5): 872-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12644843

ABSTRACT

The alpha(2)-adrenoceptor antagonist idazoxan may improve motor symptoms in Parkinson's disease and experimental Parkinsonism. We studied the effect of idazoxan on haloperidol-induced catalepsy in rats, an animal model of the drug-induced extrapyramidal side effects in man. Catalepsy was induced by a subcutaneous (s.c.) injection of haloperidol (1 mg/kg) and measured by the bar test for a maximum of 5 min. At 3 h after haloperidol, rats were given 0.16-5.0 mg/kg s.c. idazoxan, and descent latency was measured 1 h later. Idazoxan potently reversed haloperidol-induced catalepsy with an ED(50) of 0.25 mg/kg. This effect was mimicked by the selective alpha(2)-adrenoceptor antagonist RS-15385-197 (0.3 and 1 mg/kg orally). We assessed how dopaminergic mechanisms were involved in the anticataleptic effect of idazoxan by studying its effect on dopamine (DA) release in the striatum, with the microdialysis technique in conscious rats. Idazoxan (0.3 and 2.5 mg/kg) had no effect on extracellular DA and did not modify the rise of extracellular DA induced by haloperidol, indicating that changes of striatal DA release were not involved in the reversal of catalepsy. The anticataleptic effect of 2.5 mg/kg idazoxan (haloperidol+vehicle 288+/-8 s, haloperidol+idazoxan 47+/-22 s) was attenuated in rats given an intraventricular injection of 150 microg of the serotonin (5-HT) neurotoxin 5,7-dihydroxytryptamine (haloperidol+vehicle 275+/-25 s, haloperidol+idazoxan 137+/-28 s). The 5-HT(1A) receptor antagonist WAY100 635 (0.1 mg/kg s.c.) did not affect the anticataleptic effect of idazoxan. The results suggest that idazoxan reversed haloperidol-induced catalepsy by a mechanism involving blockade of alpha(2)-adrenoceptors and, at least in part, 5-HT neurons.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists , Catalepsy/drug therapy , Dopamine/metabolism , Idazoxan/therapeutic use , Serotonin/metabolism , Catalepsy/chemically induced , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , Haloperidol/toxicity , Idazoxan/pharmacology , Neurons/drug effects , Neurons/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Serotonin/metabolism , Receptors, Serotonin, 5-HT1
15.
Free Radic Biol Med ; 32(5): 394-405, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11864779

ABSTRACT

Oxidative stress is a primary pathogenesis in the brain, which is particularly vulnerable to oxidative stress. Maintenance of astrocyte functions under oxidative stress is essential to prevent neuronal injuries and to recover neuronal functions in various pathologic conditions. Imidazoline drugs have affinities for imidazoline receptors, which are highly distributed in the brain, and have been shown to be neuroprotective. This study presented the protective effects of several imidazoline drugs against oxidative cytotoxicity, in primary cultures of astrocytes. Imidazoline drugs, such as idazoxan, guanabenz, guanfacine, BU224, and RS-45041-190, showed protective effects against naphthazarin-induced oxidative cytotoxicity, as evidenced by LDH release and Hoechst 33342/propidium iodide staining. The imidazoline drugs stabilized lysosomes and inhibited naphthazarin-induced lysosomal destabilization, as evidenced by acridine orange relocation. Guanabenz inhibited, the leakage of lysosomal cathepsin D to cytosol, the decreased mitochondrial potential, and the release of mitochondrial cytochrome c, which were induced by naphthazarin. The lysosomal destabilization by oxidative stress and other apoptotic signals and subsequent cathepsin D leakage to the cytosol can induce apoptotic changes of mitochondria and eventually cell death. Therefore, lysosomal stabilization by imidazoline drugs may be ascribed to their protective effects against oxidative cytotoxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Astrocytes/metabolism , Cell Division/drug effects , Imidazoles/therapeutic use , Lysosomes/enzymology , Naphthoquinones/pharmacology , Acridine Orange , Adrenergic alpha-Antagonists/therapeutic use , Animals , Animals, Newborn , Antineoplastic Agents/adverse effects , Astrocytes/drug effects , Cathepsin D/metabolism , Cell Line , Cerebral Cortex/metabolism , Cytochrome c Group/metabolism , Fish Venoms , Guanabenz/therapeutic use , Guanfacine/therapeutic use , HIV Protease Inhibitors/pharmacology , Idazoxan/therapeutic use , Indoles/therapeutic use , Isoindoles , L-Lactate Dehydrogenase/metabolism , Ligands , Lysosomes/drug effects , Membrane Potentials/drug effects , Mitochondria/physiology , Naphthoquinones/adverse effects , Neuroglia/metabolism , Oxidation-Reduction , Pepstatins/pharmacology , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Receptors, Drug
16.
Mov Disord ; 16(4): 708-13, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11481696

ABSTRACT

Dyskinesia is a frequent and disabling side effect in patients with Parkinson's disease treated with chronic dopa-therapy. Preclinical data in the 1-methyl-4-phenyl-1,2,3,6,-tetrahydropyridine (MPTP) monkey suggest that alpha-2 antagonists may reduce dihydroxyphenylalanine (L-DOPA)-induced dyskinesia. We assessed, in a pilot randomised placebo-controlled study, the effects of single oral doses (10 mg, 20 mg, and 40 mg) of idazoxan, an alpha-2 antagonist, on motor parkinsonian disability and L-DOPA-induced dyskinesia following an acute oral challenge of L-DOPA in 18 patients with Parkinson's disease. The severity of L-DOPA-induced dyskinesia improved after 20 mg idazoxan pretreatment, while there was no concommittant deterioration in the antiparkinsonian response to L-DOPA. These results suggest that blocking alpha-2 receptors in patients with Parkinson's disease might improve L-DOPA-induced dyskinesia without the cost of a return of parkinsonian symptomatology. Further studies are required to assess whether this property could have potential therapeutic applications in the long-term management of dyskinetic patients with Parkinson's disease.


Subject(s)
Adrenergic alpha-Antagonists/therapeutic use , Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/drug therapy , Idazoxan/therapeutic use , Levodopa/adverse effects , Parkinson Disease/drug therapy , Administration, Oral , Adrenergic alpha-Antagonists/adverse effects , Aged , Antiparkinson Agents/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Idazoxan/adverse effects , Levodopa/therapeutic use , Male , Middle Aged , Neurologic Examination/drug effects , Pilot Projects
18.
Neurologia ; 15 Suppl 1: 38-42, 2000 Jan.
Article in Spanish | MEDLINE | ID: mdl-10723174

ABSTRACT

In order to treat frontotemporal dementia (FTD) we must first evaluate the patient's medical condition, as well as his or her social setting (caregiver, financial resources, home characteristics). Primary health-care team must receive information about the patient's disease, and the family should be informed about the disease itself and the social resources they can ask for. It is advisable to formulate a therapeutic scheme including some counsels to improve the suitability of environment, social help measures, behaviour therapy, cognitive stimulation and pharmacological treatment. Atypical antipsychotics have improved "positive symptoms" as logorrhoea, wandering, agitation and aggression, without impairing cognitive function. Selective serotonin reuptake inhibitors improve depressive symptoms, compulsions, food craving and disinhibition. A few reports suggest that idazoxan (alpha 2-noradrenergic antagonist) can improve attention, verbal fluency and planning efficiency. In some cases with "FTD and parkinsonism linked to chromosome 17" it could be justified to perform a genetic analysis to the offspring, in order to know if genetic counseling is necessary. An inflammatory reaction has been observed in brain damaged areas, and therefore antiinflammatory treatment efficacy should be investigated. It would also be interesting to look for neuroprotective agents that lessen the tau protein abnormality. All types of receptors which are involved in FTD should be identified, and then their selective agonists or antagonists could be administered in synergic combinations. We hope that all genetic alterations producing or facilitating FTD are eventually known, and harmless curative means are developed.


Subject(s)
Adrenergic alpha-Antagonists/therapeutic use , Dementia/drug therapy , Dementia/pathology , Frontal Lobe/pathology , Idazoxan/therapeutic use , Temporal Lobe/pathology , Humans , Primary Health Care
19.
Naunyn Schmiedebergs Arch Pharmacol ; 361(2): 181-6, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10685874

ABSTRACT

Treatment of Parkinson's disease with L-dopa is plagued in a majority of patients by dyskinesias. Noradrenaline/dopamine interactions are proposed on behavioral, biochemical, physiological and anatomical grounds. The aim of the study was to test the potential antidyskinetic effect of the alpha2-adrenoceptor antagonist, idazoxan, in a primate model of Parkinson's disease. Six female cynomolgus monkeys previously rendered parkinsonian by the toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and presenting an unchanged syndrome for several months were used. All responded readily to L-dopa but had developed dyskinesias which were manifested with each dose. In the first part of the study, seven doses of idazoxan (ranging from 0.25 mg/kg to 10 mg/kg, p.o.) were administered together with the vehicle or in combination with a fixed dose of L-dopa/benserazide (100/25 mg, p.o.). In the second part of the study, a fixed dose of idazoxan (7.5 mg/kg) was administered daily for 10 days and L-dopa was added to idazoxan on days 1, 4, 7 and 10. Vehicle (empty capsule) was used as control. Idazoxan, by itself (ranging from 5 mg/kg to 10 mg/kg), increased locomotor activity and improved the disability score with virtually no dyskinesias in three animals. In combination with L-dopa, idazoxan did not impair the antiparkinsonian response but significantly reduced dyskinesias in all six animals up to 65% at doses of 7.5 mg/kg and 10 mg/kg and delayed their onset, so that the "ON" state without dyskinesias was prolonged. The antidyskinetic effect of idazoxan was maintained when repeatedly administered for 10 days. On day 10, the locomotor response to L-dopa was significantly potentiated by chronic administration of idazoxan. Our results indicate that idazoxan has some antiparkinsonian effect of its own and may constitute a useful adjunct to L-dopa as it can reduce dyskinesias without impairing the relief of symptoms, this effect being maintained over time in this model.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists , Adrenergic alpha-Antagonists/therapeutic use , Antiparkinson Agents/therapeutic use , Dyskinesia, Drug-Induced/prevention & control , Idazoxan/therapeutic use , Levodopa/toxicity , Parkinson Disease/drug therapy , Animals , Dose-Response Relationship, Drug , Female , MPTP Poisoning/drug therapy , Macaca fascicularis , Motor Activity/drug effects , Norepinephrine/physiology , gamma-Aminobutyric Acid/metabolism
20.
J Neurosci Res ; 58(5): 690-6, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10561697

ABSTRACT

We examined the neuroprotective efficacy of a post-treatment with idazoxan (Idaz): an alpha2-adrenoceptor antagonist with activity at the I1- and I2-subtypes of the imidazoline receptor (I-receptor), in an experimental model of perinatal hypoxic-ischemic (HI) brain damage. Seventy-two, 7-day-old Wistar rats were subjected to permanent unilateral ligation of the common carotid artery and transient (2 hr) hypoxia (8% O(2)). The surviving animals were sub-divided into 3 groups: one "control" group received intraperitoneal (i.p.) injection of saline (Sigma; n = 21) and two "treated" groups received, 10 min post-HI, i.p. treatments with Idaz (I3: 3 mg/kg; n = 19) or (I8: 8 mg/kg; n = 20). Idaz effects were assessed by TTC-staining 72 hr post-HI for Sigma (n = 13), I3 (n = 11), and I8 (n = 12) groups and by MRI-examination 5 weeks post-HI for Sigma (n = 8), I3 (n = 8), and I8 (n = 6) groups. Total ratio of brain infarct areas were significantly (P < 0.01) different between Sigma and Idaz-treated rats: 20.9 +/- 4.0%, 35.6 +/- 5.9 % and 36.8 +/- 5.8% for Sigma, I3 and I8, respectively, when determined with TTC-staining and; 23.3 +/- 3.7%, 39.8 +/- 4.2%, and 43.2 +/- 10.1%, for Sigma, I3, and I8, respectively, when assessed by MRI. Our results suggest that Idaz, given as a post-HI treatment, does not exert neuroprotective effects but enhances the brain injury induced by focal neonatal cerebral HI. The deleterious mechanism may result from an overactivity of sympathetic tone and/or the immaturity of central I-receptors in newborn rats.


Subject(s)
Brain Ischemia/drug therapy , Idazoxan/adverse effects , Idazoxan/therapeutic use , Animals , Animals, Newborn , Brain/pathology , Brain Ischemia/pathology , Magnetic Resonance Imaging , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...