Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Acta Biochim Biophys Sin (Shanghai) ; 53(9): 1177-1188, 2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34244711

ABSTRACT

Stroke is the second leading cause of death and long-term disability worldwide, which lacks effective treatment. Perioperative stroke is associated with much higher rates of mortality and disability. The neuroprotective role of dexmedetomidine (Dex), a highly selective agonist of alpha2-adrenergic receptor, has been reported in a stroke rat model, and it was found that pretreatment of Dex before stroke could alleviate blood-brain barrier (BBB) breakdown. However, the underlying mechanisms are still unknown. As the brain endothelial cells are the main constituents of BBB and in high demand of energy, mitochondrial function of endothelial cells plays an important role in the maintenance of BBB. Given that dynamin-related protein 1 (Drp1) is a protein mediating mitochondrial fission, with mitochondrial fusion that balances mitochondrial morphology and ensures mitochondria function, the present study was designed to investigate the possible role of Drp1 in endothelial cells involved in the neuroprotective effects of Dex in ischemic stroke. Our results showed that preconditioning with Dex reduced infarction volume, alleviated brain water content and BBB damage, and improved neurological scores in middle cerebral artery occlusion rats. Meanwhile, Dex enhanced cell activity and decreased cell apoptosis in oxygen-glucose deprivation human brain microvascular endothelial cells in vitro. These protective effects of Dex were correlated with the mitochondrial morphology integrality of endothelial cells, mediated by increased phosphorylation of serine 637 in Drp1, and could be reversed by α2-adrenergic receptor antagonist Yohimbine and AMP-activated protein kinase inhibitor Compound C. These findings suggest new molecular pathways involved in the neuroprotective effects of Dex in ischemic stroke. As Dex is routinely used as a sedative drug clinically, our findings provide molecular evidence that it has perioperative neuroprotection from ischemic stroke.


Subject(s)
Blood-Brain Barrier/metabolism , Dexmedetomidine/pharmacology , Dynamins/metabolism , Ischemic Stroke/drug therapy , Mitochondria/metabolism , Neuroprotective Agents/pharmacology , Adenylate Kinase/antagonists & inhibitors , Adenylate Kinase/metabolism , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Animals , Blood-Brain Barrier/drug effects , Cell Line , Cytokines/metabolism , Dexmedetomidine/therapeutic use , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans , Indole Alkaloids/pharmacology , Indole Alkaloids/therapeutic use , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Ischemic Stroke/etiology , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Male , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Dynamics/drug effects , Neuroprotective Agents/therapeutic use , Phosphorylation/drug effects , Rats, Sprague-Dawley , Zonula Occludens-1 Protein/metabolism
2.
Molecules ; 26(13)2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34201675

ABSTRACT

The complex pathophysiology of depression, together with the limits of currently available antidepressants, has resulted in the continuous quest for alternative therapeutic strategies. Numerous findings suggest that pharmacological blockade of α2-adrenoceptor might be beneficial for the treatment of depressive symptoms by increasing both norepinephrine and serotonin levels in certain brain areas. Moreover, the antidepressant properties of 5-HT7 receptor antagonists have been widely demonstrated in a large set of animal models. Considering the potential therapeutic advantages in targeting both α2-adrenoceptors and 5-HT7 receptors, we designed a small series of arylsulfonamide derivatives of (dihydrobenzofuranoxy)ethyl piperidines as dually active ligands. Following green chemistry principles, the designed compounds were synthesized entirely using a sustainable mechanochemical approach. The identified compound 8 behaved as a potent α2A/5-HT7 receptor antagonist and displayed moderate-to-high selectivity over α1-adrenoceptor subtypes and selected serotonin and dopaminergic receptors. Finally, compound 8 improved performance of mice in the forced swim test, displaying similar potency to the reference drug mirtazapine.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/chemistry , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Antidepressive Agents/pharmacology , Depression/drug therapy , Motor Activity/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Receptors, Serotonin/metabolism , Adrenergic alpha-2 Receptor Antagonists/chemical synthesis , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Animals , Antidepressive Agents/therapeutic use , Behavior Rating Scale , Depression/physiopathology , HEK293 Cells , Humans , Ligands , Male , Mice , Mirtazapine/pharmacology , Mirtazapine/therapeutic use , Norepinephrine/metabolism , Piperidines/chemistry , Rats , Receptors, Serotonin/genetics , Serotonin/metabolism , Swimming
3.
J Pharmacol Exp Ther ; 375(1): 1-9, 2020 10.
Article in English | MEDLINE | ID: mdl-32665319

ABSTRACT

Treatment of fibromyalgia is an unmet medical need; however, its pathogenesis is still poorly understood. In a series of studies, we have demonstrated that some pharmacological treatments reverse generalized chronic pain but do not affect the lack of morphine analgesia in the intermittent cold stress (ICS)-induced fibromyalgia-like pain model in mice. Here we report that repeated intraperitoneal treatments with mirtazapine, which is presumed to disinhibit 5-hydroxytriptamine (5-HT) release and activate 5-HT1 receptor through mechanisms of blocking presynaptic adrenergic α2 and postsynaptic 5-HT2 and 5-HT3 receptors, completely reversed the chronic pain for more than 4 to 5 days after the cessation of treatments. The repeated mirtazapine treatments also recovered the morphine analgesia after the return of nociceptive threshold to the normal level. The microinjection of small interfering RNA (siRNA) adrenergic α2a receptor (ADRA2A) into the habenula, which showed a selective upregulation of α2 receptor gene expression after ICS, reversed the hyperalgesia but did not recover the morphine analgesia. However, both reversal of hyperalgesia and recovery of morphine analgesia were observed when siRNA ADRA2A was administered intracerebroventricularly. As the habenular is reported to be involved in the emotion/reward-related pain and hypoalgesia, these results suggest that mirtazapine could attenuate pain and/or augment hypoalgesia by blocking the habenular α2 receptor after ICS. The recovery of morphine analgesia in the ICS model, on the other hand, seems to be mediated through a blockade of α2 receptor in unidentified brain regions. SIGNIFICANCE STATEMENT: This study reports possible mechanisms underlying the complete reversal of hyperalgesia and recovery of morphine analgesia by mirtazapine, a unique antidepressant with adrenergic α2 and serotonergic receptor antagonist properties, in a type of intermittently repeated stress (ICS)-induced fibromyalgia-like pain model. Habenula, a brain region which is related to the control of emotional pain, was found to play key roles in the antihyperalgesia, whereas other brain regions appeared to be involved in the recovery of morphine analgesia in the ICS model.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Antidepressive Agents/therapeutic use , Chronic Pain/drug therapy , Fibromyalgia/drug therapy , Hyperalgesia/prevention & control , Mirtazapine/therapeutic use , Morphine/pharmacology , Adrenergic alpha-2 Receptor Antagonists/administration & dosage , Animals , Antidepressive Agents/administration & dosage , Disease Models, Animal , Gene Expression Regulation/drug effects , Injections, Intraventricular , Injections, Spinal , Male , Mice , Mice, Inbred C57BL , Mirtazapine/administration & dosage , Morphine/administration & dosage , Morphine/therapeutic use , Pain Measurement , Receptors, Adrenergic, alpha-2/genetics
4.
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
5.
Cogn Behav Ther ; 47(5): 351-371, 2018 09.
Article in English | MEDLINE | ID: mdl-29448886

ABSTRACT

The alpha-2 adrenergic receptor antagonist, yohimbine, can facilitate fear extinction in animals and humans. One potential mechanism is increased noradrenergic activity and associated arousal in the presence of conditioned stimuli. Accordingly, yohimbine might augment prolonged exposure (PE) therapy for posttraumatic stress disorder (PTSD), where heightened exposure-oriented arousal is a theorized driver and empirical predictor of treatment success. A double-blind placebo-controlled randomized trial (NCT 01031979) piloted yohimbine augmentation in 26 males with combat-related PTSD. Participants were given one-time dose of yohimbine or placebo prior to the first imaginal exposure. Subsequently, both arms completed standard PE. The primary outcome was trauma-cued heart-rate reactivity a week after the drug/exposure visit, a highly specified, objective measure sensitive to incremental change. Secondary outcomes included arousal during the drug/exposure visit and slope of distress, PTSD, and depression over the course of PE. Consistent with hypothesis, yohimbine led to higher objective and subjective arousal during the drug/exposure visit and to lower trauma-cued heart-rate reactivity one-week later. One dose of yohimbine also led to greater between-session habituation and more rapid improvement on depression, but not PTSD, over the course of care. Results of this controlled pilot indicate support for continued investigation of yohimbine-augmented exposure therapy for PTSD.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Implosive Therapy , Stress Disorders, Post-Traumatic/therapy , Yohimbine/therapeutic use , Adolescent , Adult , Combined Modality Therapy , Double-Blind Method , Fear , Humans , Male , Middle Aged , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/psychology , Treatment Outcome , Veterans/psychology , Young Adult
6.
Georgian Med News ; (283): 148-154, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30516513

ABSTRACT

An in-depth study of the pharmacological properties of 4-[4-oxo-(4h)-quinazolin-3-yl]-benzoic acid as an analgesic agent established that it had a sufficiently high analgesic effect on models of somatic and neuropathic pain syndromes. Study objective was to study the mechanisms of analgesic action of PK-66 compound in rats using the pharmacological analysis. We evaluated the mechanisms of analgesic effect of PK-66 (1 mg/kg, intraperitoneal) compound on the thermal irritation model on Hours 1, 2, 4 and 6 after administration of study compounds. To evaluate the mechanisms of PK-66 compound pain killing, we determined the changes in its efficacy against the effects of pharmacological analyzers - Naloxon, Tramadolum, Clophelinum (Clonidine), Yohimbine, Noraepinephrine, Reserpinum, Chlorpromazine (Aminazin), Levodopa, Diazepam, and Memantine). The anti-nociceptive effect of PK-66 compound was virtually unchanged during all study terms with underlying administration of Naloxon, an opioid receptor antagonist. The results of administration of Reserpinum in rats and the concomitant administration of Noradrenaline, Clophelinum, Yohimbine and quinazoline derivative demonstrated that the adrenergic system, in particular alpha-2 adrenergic receptors, was involved in the mechanisms of PK-66 activity. Changes in the PK-66 compound effect with underlying previous administration of Levodopa and Chlorpromazine suggested that the dopaminergic system was unquestionably involved in the analgesic activity of the compound. Further study of the involvement of inhibitory and exhilarating amino acids, GABA and glutamate, showed that administration of Diazepam potentiated and extended the PK-66 analgesic effect on the thermal nociception models throughout the experiment. At the same time, increased antinociception with underlying Memantine administration preceding PK-66 was observed only in the first hours of the experiment. Therefore, the studies conducted have shown that the adrenergic system, in particular alpha-2 adrenergic receptors, dopaminergic and GABAergic systems, is involved in the mechanisms of analgesic action of 4-[4-oxo-(4h) -quinazolin-3-yl]-benzoic acid (PK-66) without any effect of PK-66 on opioid receptors.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Analgesics/therapeutic use , Benzoates/therapeutic use , Dopamine D2 Receptor Antagonists/therapeutic use , Hyperalgesia/drug therapy , Pain/drug therapy , Quinazolines/therapeutic use , Administration, Oral , Adrenergic alpha-2 Receptor Antagonists/chemistry , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Analgesics/chemistry , Analgesics/pharmacology , Animals , Benzoates/pharmacology , Dopamine D2 Receptor Antagonists/chemistry , Dopamine D2 Receptor Antagonists/pharmacology , Drug Evaluation, Preclinical , Hot Temperature , Injections, Intraperitoneal , Male , Quinazolines/chemistry , Quinazolines/pharmacology , Rats, Wistar
7.
Clin Auton Res ; 27(6): 407-410, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28674870

ABSTRACT

PURPOSE: To explore the relationship between antipsychotic-associated antagonism of alpha2-adrenergic receptors and resting heart rate in individuals with schizophrenia. METHODS: Thirty-one inpatients treated with antipsychotics were included in this exploratory analysis. Antipsychotic doses were converted to haloperidol equivalents for alpha2-adrenergic receptor antagonism. Resting heart rate was measured with the patient in the seated upright posture. RESULTS: After controlling for confounding variables, the relationship between alpha2-adrenergic receptor antagonism and resting heart rate demonstrated a positive linear effect (P = 0.002) as well as a nonlinear effect that accounted for an additional 14% of the variability in resting heart rate (P = 0.005). CONCLUSION: The observed inverted-U relationship between alpha2-adrenergic receptor antagonism and resting heart rate can possibly be attributed to an altered response of beta1-adrenergic receptors to increased norepinephrine release. Further investigations are required to confirm this exploratory finding, taking into account additional variables that include other receptors which either directly or indirectly influence heart rate. CLINICALTRIALS. GOV IDENTIFIER: NCT01392885.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Antipsychotic Agents/therapeutic use , Heart Rate/drug effects , Psychotic Disorders/drug therapy , Schizophrenia/drug therapy , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Adult , Antipsychotic Agents/pharmacology , Female , Heart Rate/physiology , Humans , Male , Psychotic Disorders/physiopathology , Schizophrenia/physiopathology
8.
Int J Mol Sci ; 18(12)2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29206213

ABSTRACT

Oxaliplatin is a widely used chemotherapy agent, but induces serious peripheral neuropathy. Duloxetine is a dual reuptake inhibitor of serotonin and norepinephrine, and is shown to be effective against pain. However, whether and how duloxetine can attenuate oxaliplatin-induced allodynia in rodents is not clearly understood. A single injection of oxaliplatin (6 mg/kg, intraperitoneal; i.p.) induced a cold and mechanical allodynia, which was assessed by acetone and von Frey filament tests, respectively. When significant allodynic signs were observed, three different doses of duloxetine (10, 30, and 60 mg/kg, i.p.) were injected. Administration of 30 and 60 mg/kg of duloxetine significantly reduced the allodynia, whereas 10 mg/kg did not. By using an in vivo extracellular recording method, we further confirmed that 30 mg/kg of duloxetine could significantly inhibit the hyperexcitability of spinal wide dynamic range (WDR) cells. The anti-allodynic effect of duloxetine was completely blocked by an intrathecal injection of phentolamine (non-selective α-adrenergic receptor antagonist, 20 µg), or prazosin (α1-adrenergic receptor antagonists, 10 µg); however, idazoxan (α2-adrenergic receptor antagonist, 10 µg) did not block it. In conclusion, we suggest that duloxetine may have an effective protective action against oxaliplatin-induced neuropathic pain and spinal hyperexcitability, which is mediated by spinal α1-adrenergic receptors.


Subject(s)
Duloxetine Hydrochloride/therapeutic use , Neuralgia/chemically induced , Neuralgia/prevention & control , Neurons/drug effects , Neurons/metabolism , Organoplatinum Compounds/toxicity , Spinal Cord/cytology , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Animals , Male , Mice , Mice, Inbred C57BL , Oxaliplatin , Rats , Rats, Sprague-Dawley
9.
Int J Mol Sci ; 19(1)2017 Dec 21.
Article in English | MEDLINE | ID: mdl-29267189

ABSTRACT

Locus coeruleus-noradrenergic system dysfunction is known to contribute to the progression of Alzheimer's disease (AD). Besides a variety of reports showing the involvement of norepinephrine and its receptor systems in cognition, amyloid ß (Aß) metabolism, neuroinflammation, and neurogenesis, little is known about the contribution of the specific receptors to these actions. Here, we investigated the neurogenic and neuroprotective properties of a new α2 adrenoblocker, mesedin, in astroglial primary cultures (APC) from C57BL/6 and 3×Tg-AD mice. Our results demonstrate that mesedin rescues neuronal precursors and young neurons, and reduces the lactate dehydrogenase (LDH) release from astroglia under hypoxic and normoxic conditions. Mesedin also increased choline acetyltransferase, postsynaptic density marker 95 (PSD95), and Aß-degrading enzyme neprilysin in the wild type APC, while in the 3×Tg-AD APC exposed to glutamate, it decreased the intracellular content of Aß and enhanced the survival of synaptophysin-positive astroglia and neurons. These effects in APC can at least partially be attributed to the mesedin's ability of increasing the expression of Interleukine(IL)-10, which is a potent anti-inflammatory, neuroprotective neurogenic, and Aß metabolism enhancing factor. In summary, our data identify the neurogenic, neuroprotective, and anti-amyloidogenic action of mesedin in APC. Further in vivo studies are needed to estimate the therapeutic value of mesedin for AD.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/pharmacology , Amyloid beta-Peptides/antagonists & inhibitors , Astrocytes/drug effects , Dioxanes/pharmacology , Dioxanes/therapeutic use , Neurogenesis/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Thiazoles/pharmacology , Thiazoles/therapeutic use , Adrenergic alpha-2 Receptor Antagonists/chemistry , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Alzheimer Disease/drug therapy , Animals , Astrocytes/cytology , Biomarkers, Pharmacological/analysis , Cell Survival/drug effects , Dioxanes/chemistry , Glutamic Acid/metabolism , Hypoxia/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Primary Cell Culture , Thiazoles/chemistry
10.
J Pharmacol Exp Ther ; 358(3): 483-91, 2016 09.
Article in English | MEDLINE | ID: mdl-27418171

ABSTRACT

It has been hypothesized that α2-adrenoceptors (α2-ARs) may be involved in the pathomechanism of colitis; however, the results are conflicting because both aggravation and amelioration of colonic inflammation have been described in response to α2-AR agonists. Therefore, we aimed to analyze the role of α2-ARs in acute murine colitis. The experiments were carried out in wild-type, α2A-, α2B-, and α2C-AR knockout (KO) C57BL/6 mice. Colitis was induced by dextran sulfate sodium (DSS, 2%); alpha2-AR ligands were injected i.p. The severity of colitis was determined both macroscopically and histologically. Colonic myeloperoxidase (MPO) and cytokine levels were measured by enzyme-linked immunosorbent assay and proteome profiler array, respectively. The nonselective α2-AR agonist clonidine induced a modest aggravation of DSS-induced colitis. It accelerated the disease development and markedly enhanced the weight loss of animals, but did not influence the colon shortening, tissue MPO levels, or histologic score. Clonidine induced similar changes in α2B- and α2C-AR KO mice, whereas it failed to affect the disease activity index scores and caused only minor weight loss in α2A-AR KO animals. In contrast, selective inhibition of α2A-ARs by BRL 44408 significantly delayed the development of colitis; reduced the colonic levels of MPO and chemokine (C-C motif) ligand 3, chemokine (C-X-C motif) ligand 2 (CXCL2), CXCL13, and granulocyte-colony stimulating factor; and elevated that of tissue inhibitor of metalloproteinases-1. In this work, we report that activation of α2-ARs aggravates murine colitis, an effect mediated by the α2A-AR subtype, and selective inhibition of these receptors reduces the severity of gut inflammation.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/pharmacology , Colitis/chemically induced , Colitis/drug therapy , Dextran Sulfate/pharmacology , Intestines/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Animals , Clonidine/pharmacology , Clonidine/therapeutic use , Colitis/metabolism , Colitis/physiopathology , Drinking/drug effects , Female , Gene Knockout Techniques , Imidazoles/pharmacology , Imidazoles/therapeutic use , Intestinal Mucosa/metabolism , Intestines/pathology , Isoindoles/pharmacology , Isoindoles/therapeutic use , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Receptors, Adrenergic, alpha-2/deficiency , Receptors, Adrenergic, alpha-2/genetics
11.
Rheumatology (Oxford) ; 53(5): 948-52, 2014 May.
Article in English | MEDLINE | ID: mdl-24489014

ABSTRACT

OBJECTIVES: Our primary purpose was to evaluate the efficacy of the high-potency α2C-adrenoceptor antagonist ORM-12741 in the attenuation of a cold-induced reduction in finger blood flow and temperature in patients with RP secondary to SSc. Secondary objectives were to assess safety and tolerability. METHODS: This was a phase IIa, randomized, double-blind, crossover, single-dose, placebo-controlled, single-centre study. Patients attended five times: initial screening, treatment visits 1-3 (each at least 1 week apart) and 1-2 weeks after the last treatment. At each treatment visit, each subject received a single oral dose of 30 mg or 100 mg of ORM-12741 or placebo. Thirty minutes later the subject underwent a cold challenge. Blood flow to the fingers was assessed by three methods [temperature by probe, laser Doppler imaging (LDI) and infrared thermography] performed before, during and after the cold challenge. RESULTS: Twelve patients (10 female, mean age 58 years) were included. The area under the rewarming curve (LDI) of the right index finger (arbitrary flux units × time) was lower for both 30 mg (P = 0.043) and 100 mg (P = 0.025) of ORM-12741 compared with placebo, indicating delayed reperfusion. The time to 70% temperature recovery (middle finger probe) was longer with active than placebo treatment: mean (s.d.) values for placebo, 30 mg of ORM-12741 and 100 mg of ORM-12741 were 21.4 min (12.4), 25.7 min (12.2) and 26.9 min (13.9), respectively. Overall ORM-12741 was well tolerated. CONCLUSION: ORM-12741 did not expedite recovery from a cold challenge in the fingers of patients with SSc. TRIAL REGISTRATION: https://www.clinicaltrialsregister.eu/; no. 2010-024005-13.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/pharmacology , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Cold Temperature/adverse effects , Raynaud Disease/etiology , Raynaud Disease/prevention & control , Receptors, Adrenergic, alpha-2/drug effects , Scleroderma, Systemic/complications , Adrenergic alpha-2 Receptor Antagonists/adverse effects , Adult , Aged , Benzofurans/adverse effects , Benzofurans/pharmacology , Benzofurans/therapeutic use , Body Temperature/drug effects , Body Temperature/physiology , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Epinephrine/blood , Female , Fingers/blood supply , Humans , Laser-Doppler Flowmetry , Male , Middle Aged , Norepinephrine/blood , Quinolizidines/adverse effects , Quinolizidines/pharmacology , Quinolizidines/therapeutic use , Raynaud Disease/physiopathology , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Thermography , Treatment Outcome
12.
Psychother Psychosom ; 81(1): 29-37, 2012.
Article in English | MEDLINE | ID: mdl-22116378

ABSTRACT

BACKGROUND: Research suggests that yohimbine hydrochloride (YOH), a noradrenaline agonist, can facilitate fear extinction. It is thought that the mechanism of enhanced emotional memory is stimulated through elevated noradrenaline levels. This randomized placebo-controlled trial examined the potential exposure-enhancing effects of YOH in a clinical sample of participants meeting DSM-IV criteria for a specific phobia (fear of flying). METHODS: Sixty-seven participants with fear of flying were randomized to 4 sessions of virtual reality exposure therapy (VRET) combined with YOH (10 mg), or 4 sessions of VRET combined with a placebo. Treatment consisted of 4 weekly 1-hour exposure sessions consisting of two 25-minute virtual flights. At pre- and post- treatment, fear of flying was assessed. The YOH or placebo capsules were administered 1 h prior to exposures. The manipulation of the noradrenaline activity was confirmed by salivary α-amylase (sAA) samples taken pre-, during and post-exposure. RESULTS: Forty-eight participants completed treatment. Manipulation of noradrenaline levels with YOH was successful, with significantly higher levels of sAA in the YOH group when entering exposure. Results showed that both groups improved significantly from pre- to post-treatment with respect to anxiety reduction. However, although the manipulation of noradrenaline activity was successful, there was no evidence that YOH enhanced outcome. CONCLUSIONS: Participants improved significantly on anxiety measures independently of drug condition, after 4 sessions of VRET. These data do not support the initial findings of exposure-enhancing effects of YOH in this dosage in clinical populations.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Aerospace Medicine , Cognitive Behavioral Therapy , Extinction, Psychological/drug effects , Phobic Disorders/therapy , Yohimbine/therapeutic use , Adolescent , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Adult , Aged , Analysis of Variance , Combined Modality Therapy , Diagnostic and Statistical Manual of Mental Disorders , Fear/psychology , Humans , Implosive Therapy/methods , Middle Aged , Norepinephrine/agonists , Norepinephrine/metabolism , Phobic Disorders/drug therapy , Phobic Disorders/psychology , Placebos , Salivary alpha-Amylases/analysis , Surveys and Questionnaires , Treatment Outcome , User-Computer Interface , Yohimbine/pharmacology , Young Adult
13.
Med Clin North Am ; 106(1): 81-97, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34823736
14.
Epilepsy Behav ; 22(2): 165-77, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21813330

ABSTRACT

The postictal state is generally followed by antinociception. It is known that connections between the dorsal raphe nucleus, the periaqueductal gray matter, and the locus coeruleus, an important noradrenergic brainstem nucleus, are involved in the descending control of ascending nociceptive pathways. The aim of the present study was to determine whether noradrenergic mechanisms in the locus coeruleus are involved in postictal antinociception. Yohimbine (an α(2)-receptor antagonist) or propranolol (a ß-receptor antagonist) was microinjected unilaterally into the locus coeruleus, followed by intraperitoneal administration of pentylenetetrazole (PTZ), a noncompetitive antagonist that blocks GABA-mediated Cl(-) influx. Although the administration of both yohimbine and propranolol to the locus coeruleus/subcoeruleus area resulted in a significant decrease in tonic or tonic-clonic seizure-induced antinociception, the effect of yohimbine restricted to the locus coeruleus was more distinct compared with that of propranolol, possibly because of the presynaptic localization of α(2)-noradrenergic receptors in locus coeruleus neurons. These effects were related to the modulation of noradrenergic activity in the locus coeruleus. Interestingly, microinjections of noradrenaline into the locus coeruleus also decrease the postictal antinociception. The present results suggest that the mechanism underlying postictal antinociception involves both α(2)- and ß-noradrenergic receptors in the locus coeruleus, although the action of noradrenaline on these receptors causes a paradoxical effect, depending on the nature of the local neurotransmission.


Subject(s)
Adrenergic Neurons/drug effects , Locus Coeruleus/cytology , Norepinephrine/pharmacology , Pain Threshold/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Seizures/physiopathology , Synaptic Transmission/drug effects , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Routes , Drug Administration Schedule , Drug Interactions , Locus Coeruleus/drug effects , Male , Microinjections , Pain Measurement/drug effects , Pentylenetetrazole/toxicity , Propranolol/pharmacology , Rats , Rats, Wistar , Reaction Time/drug effects , Seizures/chemically induced , Seizures/drug therapy , Yohimbine/pharmacology , Yohimbine/therapeutic use
15.
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
16.
J Am Vet Med Assoc ; 238(9): 1159-67, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21529238

ABSTRACT

OBJECTIVE: To compare efficacy and cardiorespiratory effects of dexmedetomidine and ketamine in combination with butorphanol, hydromorphone, or buprenorphine (with or without reversal by atipamezole) in dogs undergoing castration. DESIGN: Prospective, randomized, split-plot, blinded study. ANIMALS: 30 healthy client-owned sexually intact male dogs. PROCEDURES: Dogs (n = 10 dogs/group) were assigned to receive dexmedetomidine (15 µg/kg [6.82 µg/lb]) and ketamine (3 mg/kg [1.36 mg/lb]) with butorphanol (0.2 mg/kg [0.09 mg/lb]; DKBut), the same dosages of dexmedetomidine and ketamine with hydromorphone (0.05 mg/kg [0.023 mg/lb]; DKH), or the same dosages of dexmedetomidine and ketamine with buprenorphine (40 µg/kg [18.18 µg/lb]; DKBup). All drugs were administered as a single IM injection for induction and maintenance of anesthesia for castration. At conclusion of the surgery, 5 dogs in each treatment group received atipamezole (150 µg/kg [68.18 µg/lb], IM), and the remainder received saline (0.9% NaCl) solution IM. Cardiorespiratory variables and quality of anesthesia were assessed. Supplemental isoflurane was administered to the dogs when anesthesia was considered inadequate during surgery. RESULTS: All drug combinations rapidly induced anesthesia. Dogs were intubated within 10 minutes after injection. Supplemental isoflurane was needed during surgery in 1, 3, and 4 dogs in the DKBup, DKBut, and DKH groups, respectively. Dogs that received atipamezole had a significantly shorter recovery time. Some dogs in each group had bradycardia and hypoxemia with hypertension. CONCLUSIONS AND CLINICAL RELEVANCE: DKBup was the most suitable injectable anesthetic combination used. Recovery was shortened by IM administration of atipamezole. There were minimal adverse effects in all groups.


Subject(s)
Analgesics, Opioid/pharmacology , Dexmedetomidine/pharmacology , Ketamine/pharmacology , Orchiectomy/veterinary , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Analgesics, Opioid/administration & dosage , Anesthesia, General/veterinary , Anesthetics, Dissociative/administration & dosage , Anesthetics, Dissociative/pharmacology , Animals , Dexmedetomidine/administration & dosage , Dogs , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacology , Imidazoles/therapeutic use , Ketamine/administration & dosage , Male
17.
Inflammation ; 44(1): 80-90, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32789555

ABSTRACT

Local joint inflammation plays an important role in the pathogenesis of temporomandibular joint (TMJ) osteoarthrosis (TMJOA). Yohimbine, an alpha-2 adrenergic receptor antagonist, possesses anti-inflammatory properties; however, the ability of Yohimbine to protect against TMJOA-associated chondrocyte inflammation remains unclear. We conducted in vitro and in vivo analyses to investigate whether Yohimbine could ameliorate TMJOA-induced chondrocyte inflammation and to elucidate the mechanisms involved. Chondrocytes of TMJOA mice were stimulated with interleukin (IL)-1ß or noradrenaline (NE), and the resulting production of inflammation-related factors was evaluated in the presence or absence of Yohimbine. Furthermore, two TMJOA mouse models were treated with Yohimbine and the therapeutic effect was quantified. NE (10-6 M) triggered inflammatory cytokine secretion by TMJ chondrocytes, and Yohimbine suppressed IL-1ß- or NE-induced IL-6 upregulation in TMJ chondrocytes with the nuclear factor (NF)-κB pathway inhibition. Yohimbine also ameliorated cartilage destruction in the TMJOA models. Interestingly, αmpT, a tyrosine hydroxylase inhibitor, reversed the effects of Yohimbine by activating the NF-κB pathway. Collectively, these findings show that Yohimbine ameliorated TMJ chondrocyte inflammation and the suppression of NF-κB pathway contributes to this effect.


Subject(s)
Chondrocytes/drug effects , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Temporomandibular Joint Disorders/drug therapy , Temporomandibular Joint/drug effects , Yohimbine/therapeutic use , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Animals , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/pathology , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Signal Transduction/physiology , Temporomandibular Joint/metabolism , Temporomandibular Joint/pathology , Temporomandibular Joint Disorders/metabolism , Temporomandibular Joint Disorders/pathology , Yohimbine/pharmacology
18.
Mov Disord ; 25(13): 2084-93, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20824735

ABSTRACT

Reduction in the antiparkinsonian benefit of levodopa is a major complication of long-term levodopa treatment in advanced Parkinson's disease (PD). Such loss of benefit arises because of reduced duration of action and appearance of disabling dyskinesia. We assess the potential of the α(2) adrenergic antagonist fipamezole to reduce motor complications in parkinsonian macaques. MPTP-lesioned macaques were treated acutely with fipamezole (10 mg/kg) alone and in combination with two doses of levodopa. Fipamezole extended both duration and quality of antiparkinsonian action of levodopa. Duration of antiparkinsonian action, on time, was increased by up to 75% while "good-quality" on time, i.e., that not associated with disabling dyskinesia, was increased by up to 98%. Combination of fipamezole with the lower dose of levodopa provided antiparkinsonian benefit at least equivalent to that provided by the higher dose levodopa alone. However, with the combination, antiparkinsonian benefit was of much better quality. The proportion of on time without disabling dyskinesia (79%) was significantly greater than that with high dose levodopa alone (45%). Increased duration and quality of levodopa action may represent therapeutically valuable actions of α(2) adrenergic antagonists.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Dopamine Agents/therapeutic use , Imidazoles/therapeutic use , Indans/therapeutic use , Levodopa/therapeutic use , Parkinsonian Disorders/drug therapy , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Analysis of Variance , Animals , Disability Evaluation , Disease Models, Animal , Dopamine Agents/adverse effects , Dopamine Agents/pharmacology , Drug Therapy, Combination/methods , Dyskinesia, Drug-Induced/drug therapy , Dyskinesia, Drug-Induced/etiology , Female , Imidazoles/pharmacology , Indans/pharmacology , Levodopa/adverse effects , Levodopa/pharmacology , Macaca fascicularis , Male , Motor Activity/drug effects , Time Factors
19.
Vet Clin North Am Equine Pract ; 26(3): 603-17, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21056302

ABSTRACT

Identification and alleviation of visceral pain is a frequent concern for the equine owner and veterinarian. This article discusses sources, methods for identification and quantitation, and options for treatment of visceral pain in horses.


Subject(s)
Horse Diseases/therapy , Pain/veterinary , Viscera/pathology , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Analgesia/veterinary , Analgesics/therapeutic use , Analgesics, Opioid/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Combined Modality Therapy , Horse Diseases/diagnosis , Horses , Pain/diagnosis , Pain Management , Pain Measurement/veterinary , Viscera/drug effects , Viscera/surgery
20.
Urol Nurs ; 30(4): 247-51, 2010.
Article in English | MEDLINE | ID: mdl-20949811

ABSTRACT

Female sexual arousal disorder (FSAD) occurs when a woman experiences distress due to the inability to attain or maintain adequate vaginal lubrication. Many treatments are still under investigation, and at this time, there are no FDA-approved medications available for the treatment of FSAD. Treatment options include "off-label" medications, vaginal estrogen, compounded preparations, and over-the-counter products.


Subject(s)
Sexual Dysfunction, Physiological/prevention & control , Sexual Dysfunctions, Psychological/prevention & control , Administration, Intravaginal , Adrenergic alpha-2 Receptor Antagonists/therapeutic use , Alprostadil/therapeutic use , Estrogen Replacement Therapy , Female , Glycosides/therapeutic use , Humans , Lubricants , Minerals/therapeutic use , Nonprescription Drugs/therapeutic use , Off-Label Use , Piperazines/therapeutic use , Plant Preparations/therapeutic use , Purines/therapeutic use , Sexual Dysfunction, Physiological/etiology , Sexual Dysfunctions, Psychological/etiology , Sildenafil Citrate , Suction , Sulfones/therapeutic use , Vasodilator Agents/therapeutic use , Yohimbine/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL