Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Eur Neuropsychopharmacol ; 69: 26-46, 2023 04.
Article in English | MEDLINE | ID: mdl-36706689

ABSTRACT

To study mental illness and health, in the past researchers have often broken down their complexity into individual subsystems (e.g., genomics, transcriptomics, proteomics, clinical data) and explored the components independently. Technological advancements and decreasing costs of high throughput sequencing has led to an unprecedented increase in data generation. Furthermore, over the years it has become increasingly clear that these subsystems do not act in isolation but instead interact with each other to drive mental illness and health. Consequently, individual subsystems are now analysed jointly to promote a holistic understanding of the underlying biological complexity of health and disease. Complementing the increasing data availability, current research is geared towards developing novel methods that can efficiently combine the information rich multi-omics data to discover biologically meaningful biomarkers for diagnosis, treatment, and prognosis. However, clinical translation of the research is still challenging. In this review, we summarise conventional and state-of-the-art statistical and machine learning approaches for discovery of biomarker, diagnosis, as well as outcome and treatment response prediction through integrating multi-omics and clinical data. In addition, we describe the role of biological model systems and in silico multi-omics model designs in clinical translation of psychiatric research from bench to bedside. Finally, we discuss the current challenges and explore the application of multi-omics integration in future psychiatric research. The review provides a structured overview and latest updates in the field of multi-omics in psychiatry.


Subject(s)
Mental Disorders , Multiomics , Humans , Genomics , Proteomics/methods , Machine Learning , Mental Disorders/diagnosis , Mental Disorders/genetics , Mental Disorders/therapy
2.
BMC Neurol ; 22(1): 251, 2022 Jul 08.
Article in English | MEDLINE | ID: mdl-35804294

ABSTRACT

BACKGROUND: Eptinezumab is an anti-calcitonin gene-related peptide humanized monoclonal antibody approved for the preventive treatment of migraine in adults. The PREVAIL study demonstrated a favorable safety profile with sustained reductions in overall migraine-related burden in patients with chronic migraine (CM). This post hoc analysis aimed to examine item-level changes in the Migraine Disability Assessment (MIDAS) questionnaire over 2 years in participants with CM on eptinezumab treatment. METHODS: PREVAIL was an open-label, phase 3 trial that included 96 weeks of treatment where 128 adults received intravenous eptinezumab administered over 30 min every 12 weeks (wks) for up to 8 doses of 300 mg. MIDAS was administered at baseline, Wk12, and every 12wks thereafter. Two supplementary MIDAS items not included in the total score calculation assessed number of headache days in the past 3 months (MIDAS headache) and average headache pain severity (from 0 [none] to 10 [worst]). MIDAS total scores were summed from 5 items, each quantifying the number of days in the past 3 months with migraine-related disability. Items 1, 3, and 5 assessed absenteeism, namely how many days the patient missed work/school (Q1), household work (Q3), or family/social/leisure activities (Q5). Items 2 and 4 were measures of presenteeism, namely how many days the patient had reduced productivity in work/school (Q2) or household work (Q4). RESULTS: Mean MIDAS headache days decreased from 47.4 (baseline) to 17.1 (Wk12) and 16.3 (Wk104). The average headache pain severity score (0‒10) decreased from a mean of 7.3 (baseline) to 5.5 (Wk12) to 4.5 (Wk104). Mean MIDAS scores measuring absenteeism (Q1, 3, 5) changed from 9.7 days at baseline to 3.2 days (Wk12) and to 3.9 days (Wk104). Mean MIDAS scores measuring presenteeism (Q2, 4) at Wk12 decreased from 14.2 days at baseline to 5.2 days (Wk12, 104). Patients categorized with very severe MIDAS disability had a mean total MIDAS score of 84.8, with an average reduction of 56.7 days (Wk12), which was maintained at 32 days at Wk104. CONCLUSIONS: Long-term treatment with eptinezumab in patients with CM suggested sustained reductions in MIDAS-quantified disability, consistent with the sustained reductions in headache frequency and pain severity. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02985398 .


Subject(s)
Migraine Disorders , Adult , Antibodies, Monoclonal, Humanized/therapeutic use , Chronic Disease , Disability Evaluation , Double-Blind Method , Headache , Humans , Migraine Disorders/drug therapy , Migraine Disorders/prevention & control , Treatment Outcome
3.
Neuropsychiatr Dis Treat ; 17: 2995-3006, 2021.
Article in English | MEDLINE | ID: mdl-34588779

ABSTRACT

PURPOSE: This qualitative study explored patient perceptions of the most burdensome symptoms of major depressive disorder (MDD), the impact of symptoms on patients' daily lives, and patient expectations and experiences regarding the timing of onset of antidepressant pharmacotherapy. PATIENTS AND METHODS: Data were collected through facilitated, patient focus-group sessions in the USA between May and June 2019. Participants were adults with confirmed MDD who reported a major depressive episode within the past 2 years, for which they had received pharmacologic treatment for ≥6 weeks. The semi-structured discussion focused on the key topics of bothersome symptoms of MDD, the impact of symptoms on quality of life, and the effects of antidepressant treatment. Interviews were audio-recorded; findings were summarized using a content-analysis approach. RESULTS: Five focus-group sessions were undertaken, involving a total of 29 patients (each attended one session; mean age, 43.4 years; 72.4% female). Mean time since confirmed diagnosis of MDD was 13.1 years. The most commonly prescribed antidepressants received were bupropion (41.4% of participants), escitalopram (34.5%), and sertraline (34.5%). The most frequently reported bothersome MDD symptoms were fatigue (mentioned by 58.6% of participants), lack of motivation/loss of interest (51.7%), anxiety/panic (44.8%), sadness (41.4%), and lack of concentration/brain fog (41.4%). Socialization, family life, and work were the areas in which quality of life was most impacted. Participants expressed dissatisfaction with their antidepressant treatment. Fast symptom resolution was mentioned as a priority (defined as <1 week by 38.5% of participants and ≤1 month by 65.4%). Most participants had not experienced fast relief from their symptoms with current or previous antidepressant medications. CONCLUSION: Results of this qualitative study suggest that fatigue, anhedonia, cognitive symptoms, and anxiety are some of the most bothersome symptoms for patients with MDD and highlight the importance of obtaining rapid relief from these symptoms in order to improve outcomes and patient satisfaction with antidepressant medication.

4.
Brain Res ; 1689: 1-11, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29274875

ABSTRACT

The multimodal antidepressant vortioxetine is thought to mediate its pharmacological effects via 5-HT1A receptor agonism, 5-HT1B receptor partial agonism, 5-HT1D, 5-HT3, 5-HT7 receptor antagonism and 5-HT transporter inhibition. Here we studied vortioxetine's functional effects across species (canine, mouse, rat, guinea pig and human) in cellular assays with heterologous expression of 5-HT3A receptors (in Xenopus oocytes and HEK-293 cells) and in mouse neuroblastoma N1E-115 cells with endogenous expression of 5-HT3A receptors. Furthermore, we studied the effects of vortioxetine on activity of CA1 Stratum Radiatum interneurons in rat hippocampus slices using current- and voltage-clamping methods. The patched neurons were subsequently filled with biocytin for confirmation of 5-HT3 receptor mRNA expression by in situ hybridization. Whereas, both vortioxetine and the 5-HT3 receptor antagonist ondansetron potently antagonized 5-HT-induced currents in the cellular assays, vortioxetine had a slower off-rate than ondansetron in oocytes expressing 5-HT3A receptors. Furthermore, vortioxetine's but not ondansetron's 5-HT3 receptor antagonistic potency varied considerably across species. Vortioxetine had the highest potency at rat and the lowest potency at guinea pig 5-HT3A receptors. Finally, in 5-HT3 receptor-expressing GABAergic interneurons from the CA1 stratum radiatum, vortioxetine and ondansetron blocked depolarizations induced by superfusion of either 5-HT or the 5-HT3 receptor agonist mCPBG. Taken together, these data add to a growing literature supporting the idea that vortioxetine may inhibit GABAergic neurotransmission in some brain regions via a 5-HT3 receptor antagonism-dependent mechanism and thereby disinhibit pyramidal neurons and enhance glutamatergic signaling.


Subject(s)
Action Potentials/drug effects , Antidepressive Agents/pharmacology , Interneurons/drug effects , Pyramidal Cells/drug effects , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Vortioxetine/pharmacology , Action Potentials/physiology , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Dogs , Glutamic Acid/metabolism , Guinea Pigs , HEK293 Cells , Humans , Interneurons/metabolism , Mice , Ondansetron/pharmacology , Oocytes , Pyramidal Cells/metabolism , Rats , Receptors, Serotonin, 5-HT3/metabolism , Serotonin/metabolism , Tissue Culture Techniques , Xenopus laevis , gamma-Aminobutyric Acid/metabolism
5.
BMC Psychiatry ; 17(1): 370, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29157207

ABSTRACT

BACKGROUND: The manifestation of major depressive disorder (MDD) may include cognitive symptoms that can precede the onset of MDD and persist beyond the resolution of acute depressive episodes. However, little is known about how cognitive symptoms are experienced by MDD patients and the people around them. METHODS: In this international (Brazil, Canada, China, France, and Germany) ethnographic study, we conducted semi-structured interviews and observations of remitted as well as symptomatic MDD patients (all patients self-reported being diagnosed by an HCP and self-reported being on an antidepressant) aged 18-60 years with self-reported cognitive symptoms (N = 34). In addition, participating depressed patients' close family or friends (N = 31) were interviewed. Separately recruited from depressed participants, work colleagues (N = 21) and healthcare providers (HCPs; N = 13) of depressed individuals were interviewed. RESULTS: Key insights were that: (1) patients were generally unaware that their cognitive symptoms were linked to their depression and, instead, attributed these symptoms to negative aspects of their person (e.g., age, separate disease, laziness, exhaustion); (2) cognitive symptoms in MDD appeared to negatively impact patients' social relationships and patients' ability to handle daily tasks at work and at home; (3) patients' cognitive symptoms also impacted relationships with family members and coworkers; (4) patients' cognitive symptoms increased stress and feelings of failure, which in turn seemed to worsen the cognitive symptoms, thereby creating a destructive cycle; and (5) although HCPs recommended that patients re-engage in everyday activities to help overcome their depression, cognitive symptoms seemed to impede such functional recovery. CONCLUSIONS: Taken together, these findings highlight a negative impact of patients' cognitive symptoms on their social functioning, work performance, and quality of life on the people close to them, and consequently on the degree of functional recovery after depression.


Subject(s)
Activities of Daily Living/psychology , Cognitive Dysfunction/psychology , Depressive Disorder, Major/psychology , Family/psychology , Friends/psychology , Adolescent , Adult , Antidepressive Agents/therapeutic use , Brazil/ethnology , Canada/ethnology , China/ethnology , Cognitive Dysfunction/ethnology , Cost of Illness , Depressive Disorder, Major/ethnology , Diagnostic Self Evaluation , Ethnopsychology , Family/ethnology , Female , France/ethnology , Friends/ethnology , Germany/ethnology , Humans , Male , Middle Aged , Qualitative Research , Young Adult
6.
PLoS One ; 10(3): e0120239, 2015.
Article in English | MEDLINE | ID: mdl-25798598

ABSTRACT

The 1,5-benzodiazepine clobazam is indicated for the adjunctive treatment of seizures associated with Lennox-Gastaut syndrome in patients 2 years of age or older in the United States, and for treatment of anxiety and various forms of epilepsy elsewhere. Clobazam has been reported to exhibit different in vivo adverse effects and addiction liability profile than the classic 1,4-benzodiazepines. In this study, it was investigated whether the in vitro pharmacological properties of clobazam and its major active metabolite N-desmethylclobazam could explain some of these clinical differences. The functional properties of the two 1,5-benzodiazepines were characterized at the human γ-aminobutyric acid type A receptor (GABA(A)R) subtypes α1ß2γ(2S), α2ß2γ(2S), α3ß2γ(2S), α5ß2γ(2S) and α6ß2δ expressed in Xenopus laevis oocytes by use of two-electrode voltage-clamp electrophysiology and compared to those exhibited by the 1,4-benzodiazepine clonazepam. All three compounds potentiated GABA EC20-evoked responses through the α(1,2,3,5)ß2γ(2S) GABA(A)Rs in a reversible and concentration-dependent manner, with each displaying similar EC50 values at the four subtypes. Furthermore, the degrees of potentiation of the GABA EC20 currents through the four receptors mediated by saturating modulator concentrations did not differ substantially for any of the three benzodiazepines. The three compounds were substantially less potent (200-3900 fold) as positive allosteric modulators at the α6ß2δ GABA(A)R than at the α(1,2,3,5)ß2γ(2S) receptors. Interestingly, however, clobazam and especially N-desmethylclobazam were highly efficacious potentiators of α6ß2δ receptor signaling. Although this activity component is unlikely to contribute to the in vivo effects of clobazam/N-desmethylclobazam, the 1,5-benzodiazepine could constitute an interesting lead for novel modulators targeting this low-affinity binding site in GABAARs. In conclusion, the non-selective modulation exerted by clobazam, N-desmethylclobazam and clonazepam at the α1ß2γ(2S), α2ß2γ(2S), α3ß2γ(2S) and α5ß2γ(2S) GABA(A)Rs indicate that the observed clinical differences between clobazam and 1,4-benzodiazepines are likely to arise from factors other than their respective pharmacological properties at the GABA(A)Rs as investigated here.


Subject(s)
Benzodiazepines/metabolism , Benzodiazepines/pharmacology , Oocytes/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Xenopus laevis/genetics , Animals , Clobazam , GABA-A Receptor Agonists/metabolism , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Antagonists/metabolism , GABA-A Receptor Antagonists/pharmacology , Gene Expression , Humans
7.
PLoS One ; 9(2): e88456, 2014.
Article in English | MEDLINE | ID: mdl-24533090

ABSTRACT

Clobazam (CLB), a 1,5-benzodiazepine (BZD), was FDA-approved in October 2011 for the adjunctive treatment of seizures associated with Lennox-Gastaut syndrome (LGS) in patients 2 years and older. BZDs exert various CNS effects through allosteric modulation of GABAA receptors. The structurally distinct, 1,4-BZD clonazepam (CLN) is also approved to treat LGS. The precise mechanisms of action and clinical efficacy of both are unknown. Data show that the GABAA α1-subunit-selective compound zolpidem [ZOL] exhibits hypnotic/sedative effects. Conversely, data from knock-in mice carrying BZD binding site mutations suggest that the α2 subunit mediates anticonvulsant effects, without sedative actions. Hence, the specific pattern of interactions across the GABAA receptor complexes of BZDs might be reflected in their clinical efficacies and adverse effect profiles. In this study, GABAA-receptor binding affinities of CLB, N-desmethylclobazam (N-CLB, the major metabolite of CLB), CLN, and ZOL were characterized with native receptors from rat-brain homogenates and on cloned receptors from HEK293 cells transfected with combinations of α (α1, α2, α3, or α5), ß2, and γ2 subtypes. Our results demonstrate that CLB and N-CLB have significantly greater binding affinities for α2- vs. α1-receptor complexes, a difference not observed for CLN, for which no distinction between α2 and α1 receptors was observed. Our experiments with ZOL confirmed the high preference for α1 receptors. These results provide potential clues to a new understanding of the pharmacologic modes of action of CLB and N-CLB.


Subject(s)
Anticonvulsants/pharmacology , Benzodiazepines/pharmacology , Allosteric Site , Animals , Anticonvulsants/chemistry , Benzodiazepines/chemistry , Binding Sites , Brain/metabolism , Clobazam , Clonazepam/chemistry , Clonazepam/pharmacology , HEK293 Cells , Humans , Kinetics , Male , Mice , Mutation , Protein Binding , Pyridines/chemistry , Pyridines/pharmacology , Rats , Receptors, GABA-A/metabolism , Seizures/drug therapy , Zolpidem
8.
Neuropharmacology ; 73: 147-59, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23721744

ABSTRACT

Vortioxetine (Lu AA21004) is an investigational novel antidepressant with multimodal activity that functions as a 5-HT3, 5-HT7 and 5-HT(1D) receptor antagonist, 5-HT(1B) receptor partial agonist, 5-HT(1A) receptor agonist and inhibitor of the 5-HT transporter in vitro. Here we explore its anxiolytic and antidepressant potential in adult mice. Vortioxetine was assessed in BalB/cJ@RJ mice using the open-field and forced-swim tests (acute: p.o. 1 h, repeated: daily p.o. 21 days), and in 129S6/SvEvTac mice using the novelty suppressed feeding paradigm (acute: p.o. 1 h, sustained: daily p.o. 14 or 21 days). Fluoxetine and diazepam were controls. Acute and repeated dosing of vortioxetine produced more pronounced anxiolytic- and antidepressant-like activities than fluoxetine. Vortioxetine significantly increased cell proliferation and cell survival and stimulated maturation of immature granule cells in the subgranular zone of the dentate gyrus of the hippocampus after 21 days of treatment. After 14 days, a high dose of vortioxetine increased dendritic length and the number of dendrite intersections, suggesting that vortioxetine accelerates the maturation of immature neurons. Vortioxetine displays an antidepressant and anxiolytic profile following repeated administration associated with increased neurogenesis at several stages. Vortioxetine effects were observed at low levels of 5-HT transporter occupancy, suggesting an alternative mechanism of action to 5-HT reuptake inhibition.


Subject(s)
Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Neurogenesis/drug effects , Piperazines/pharmacology , Sulfides/pharmacology , Animals , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Diazepam/pharmacology , Drug Evaluation, Preclinical , Exploratory Behavior/drug effects , Fluoxetine/pharmacology , Immobility Response, Tonic/drug effects , Male , Mice , Motor Activity/drug effects , Vortioxetine
9.
Eur Neuropsychopharmacol ; 23(2): 133-45, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22612991

ABSTRACT

The monoaminergic network, including serotonin (5-HT), norepinephrine (NE), and dopamine (DA) pathways, is highly interconnected and has a well-established role in mood disorders. Preclinical research suggests that 5-HT receptor subtypes, including 5-HT1A, 5-HT1B, 5-HT3, and 5-HT7 receptors as well as the 5-HT transporter (SERT), may have important roles in treating depression. This study evaluated the neuropharmacological profile of Lu AA21004, a novel multimodal antidepressant combining 5-HT3 and 5-HT7 receptor antagonism, 5-HT1B receptor partial agonism, 5-HT1A receptor agonism, and SERT inhibition in recombinant cell lines. Extracellular 5-HT, NE and DA levels were evaluated in the ventral hippocampus (vHC), medial prefrontal cortex (mPFC) and nucleus accumbens (NAc) after acute and subchronic treatment with Lu AA21004 or escitalopram. The acute effects of LuAA21004 on NE and DA neuronal firing were also evaluated in the locus coeruleus (LC) and ventral tegmental area (VTA), respectively. Acute Lu AA21004 dose-dependently increased 5-HT in the vHC, mPFC and NAc. Maximal 5-HT levels in the vHC were higher than those in the mPFC. Furthermore, mPFC 5-HT levels were increased at low SERT occupancy levels. In the vHC and mPFC, but not the NAc, high Lu AA21004 doses increased NE and DA levels. Lu AA21004 slightly decreased LC NE neuronal firing and had no effect on VTA DA firing. Results are discussed in context of occupancy at 5-HT3, 5-HT1B and 5-HT1A receptors and SERT. In conclusion, Lu AA21004, acting via two pharmacological modalities, 5-HT receptor modulation and SERT inhibition, results in a brain region-dependent increase of multiple neurotransmitter concentrations.


Subject(s)
Antidepressive Agents/pharmacology , Neurons/physiology , Neurotransmitter Agents/metabolism , Piperazines/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Sulfides/pharmacology , Animals , Biogenic Monoamines/metabolism , Citalopram/pharmacology , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/metabolism , Locus Coeruleus/drug effects , Locus Coeruleus/physiology , Male , Neurons/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1B/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology , Vortioxetine
10.
Int J Neuropsychopharmacol ; 16(5): 1115-27, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23089374

ABSTRACT

The therapeutic effect of current antidepressant drugs appears after several weeks of treatment and a significant number of patients do not respond to treatment. Here, we report the effects of the multi-modal antidepressant vortioxetine (Lu AA21004), a 5-HT(3) and 5-HT(7) receptor antagonist, 5-HT(1B) receptor partial agonist, 5-HT(1A) receptor agonist and 5-HT transporter (SERT) inhibitor, on rat 5-HT neurotransmission. Using in vivo electrophysiological recordings in the dorsal raphe nucleus of anaesthetized rats, we assessed the acute and subchronic effects of vortioxetine and/or the selective 5-HT(3) receptor agonist, SR57227 or the selective 5-HT(1A) receptor agonist flesinoxan, on 5-HT neuronal firing activity. Using ex-vivo autoradiography, we correlated SERT occupancy and presumed 5-HT firing activity. The selective serotonin reuptake inhibitor, fluoxetine, was used as comparator. Importantly, the recovery of 5-HT neuronal firing was achieved after 1 d with vortioxetine and 14 d with fluoxetine. SR57227 delayed this recovery. In contrast, vortioxetine failed to alter the reducing action of 3 d treatment of flesinoxan. Acute dosing of vortioxetine inhibited neuronal firing activity more potently than fluoxetine. SR57227 prevented the suppressant effect of vortioxetine, but not of fluoxetine. In contrast, flesinoxan failed to modify the suppressant effect of vortioxetine acutely administered. Differently to fluoxetine, vortioxetine suppressed neuronal firing without saturating occupancy at the SERT. Vortioxetine produced a markedly faster recovery of 5-HT neuronal firing than fluoxetine. This is at least partly due to 5-HT(3) receptor antagonism of vortioxetine in association with its reduced SERT occupancy.


Subject(s)
Action Potentials/drug effects , Antidepressive Agents/pharmacology , Neurons/drug effects , Piperazines/pharmacology , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Serotonin/metabolism , Sulfides/pharmacology , Animals , Autoradiography , Drug Administration Schedule , Drug Delivery Systems , Electrolytes/adverse effects , Male , Piperidines/pharmacology , Raphe Nuclei/cytology , Raphe Nuclei/injuries , Rats , Rats, Sprague-Dawley , Receptors, Serotonin, 5-HT3/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Serotonin Receptor Agonists/pharmacology , Time Factors , Vortioxetine
11.
Neuropharmacology ; 63(3): 469-79, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22579928

ABSTRACT

δ-subunit containing extrasynaptic GABA(A) receptors are potential targets for modifying neuronal activity in a range of brain disorders. With the aim of gaining more insight in synaptic and extrasynaptic inhibition, we used a new positive modulator, AA29504, of δ-subunit containing GABA(A) receptors in mouse neurons in vitro and in vivo. Whole-cell patch-clamp recordings were carried out in the dentate gyrus in mouse brain slices. In granule cells, AA29504 (1 µM) caused a 4.2-fold potentiation of a tonic current induced by THIP (1 µM), while interneurons showed a potentiation of 2.6-fold. Moreover, AA29504 (1 µM) increased the amplitude and prolonged the decay of miniature inhibitory postsynaptic currents (mIPSCs) in granule cells, and this effect was abolished by Zn²âº (15 µM). AA29504 (1 µM) also induced a small tonic current (12.7 ± 3.2 pA) per se, and when evaluated in a nominally GABA-free environment using Ca²âº imaging in cultured neurons, AA29504 showed GABA(A) receptor agonism in the absence of agonist. Finally, AA29504 exerted dose-dependent stress-reducing and anxiolytic effects in mice in vivo. We propose that AA29504 potentiates δ-containing GABA(A) receptors to enhance tonic inhibition, and possibly recruits perisynaptic δ-containing receptors to participate in synaptic phasic inhibition in dentate gyrus.


Subject(s)
GABA Agents/pharmacology , GABA Agonists/pharmacology , Neurons/drug effects , Neurons/physiology , Receptors, GABA-A/physiology , Animals , Anxiety/drug therapy , Anxiety/psychology , Brain/metabolism , Calcium/metabolism , Data Interpretation, Statistical , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Electrophysiological Phenomena , Fever/etiology , GABA Agents/metabolism , Isoxazoles/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Patch-Clamp Techniques , Pyramidal Cells/drug effects , Radioligand Assay , Receptors, GABA-A/drug effects , Stress, Psychological/physiopathology , Synaptic Transmission/drug effects
12.
J Neurosci ; 32(15): 5310-20, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22496576

ABSTRACT

Dopamine neurons of the ventral tegmental area (VTA) are involved at early phases of drug addiction. Even the first in vivo dose of various abused drugs induces glutamate receptor plasticity at the excitatory synapses of these neurons. Benzodiazepines that suppress the inhibitory GABAergic interneurons in the VTA via facilitation of synaptic GABA(A) receptors have induced neuroplasticity in dopamine neurons due to this disinhibitory mechanism. Here, we have tested a non-benzodiazepine direct GABA site agonist 4,5,6,7-tetrahydroisoxazolol[4,5-c]pyridine-3-ol (THIP) (also known as gaboxadol) that acts preferentially via high-affinity extrasynaptic GABA(A) receptors. A single sedative dose of THIP (6 mg/kg) to mice induced glutamate receptor plasticity for at least 6 d after administration. Increased AMPA/NMDA receptor current ratio and increased frequency, amplitude, and rectification of AMPA receptor responses suggested persistent targeting of GluA2-lacking AMPA receptors in excitatory synapses of VTA dopamine neurons ex vivo after THIP administration. This effect was abolished in GABA(A) receptor δ(-/-) mice, which have a loss of extrasynaptic GABA(A) receptors. In behavioral experiments, we found neither acute reinforcement in intravenous self-administration sessions with THIP at relevant doses using a yoked control paradigm in mice nor in baboons using a standard paradigm for assessing drug abuse liability; nor was any place preference found after conditioning sessions with various doses of THIP but rather a persistent aversion in 6 mg/kg THIP-conditioned mice. In summary, we found that activation of extrasynaptic δ-subunit-containing GABA(A) receptors leads to glutamate receptor plasticity of VTA dopamine neurons, but is not rewarding, and, instead, induces aversion.


Subject(s)
Dopaminergic Neurons/drug effects , GABA Agonists/pharmacology , Isoxazoles/pharmacology , Reward , Substance-Related Disorders/pathology , Ventral Tegmental Area/drug effects , Animals , Behavior, Animal/drug effects , Electrophysiological Phenomena , Excitatory Postsynaptic Potentials/drug effects , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Neuronal Plasticity/drug effects , Papio , Polymerase Chain Reaction , Receptors, AMPA/drug effects , Receptors, GABA-A/genetics , Receptors, GABA-A/physiology , Self Administration , Ventral Tegmental Area/cytology
13.
Eur Neuropsychopharmacol ; 22(10): 751-60, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22406239

ABSTRACT

According to current theories on the etiopathogenesis and pathophysiology of depression, both GABAergic and monoaminergic transmitter systems are perturbed. Consequently, the present study addressed the putative antidepressant action of the sedative-hypnotic GABAA receptor agonist, gaboxadol, separately and in combination with the selective serotonin reuptake inhibitor (SSRI) escitalopram. The rat chronic mild stress model was used to test the chronic antidepressant properties of gaboxadol in this depression model. Sucrose intake used as a read-out on anhedonic-like behavior indicated that the drug response rate for gaboxadol (5 mg/kg/day, i.p.) was similar to that measured for escitalopram (5 mg/kg/day, i.p.), however, the rate increased when the two drugs were co-administered, suggesting a synergistic action. Using in vivo electrophysiological recordings in dorsal raphe nucleus (DRN) of anesthetised rats, the present results showed that one week treatment with gaboxadol (5 mg/kg/day, i.p.) or with escitalopram (5 mg/kg/day, i.p.), followed by a 24 h washout period, did not affect DRN 5-HT neuronal firing per se, but in rats treated with both drugs for one week, the firing rate of DRN 5-HT neurons was significantly increased. Immunohistochemical estimations of cell proliferation in the hippocampal dentate gyrus did not reveal any effect of gaboxadol on chronic mild stressed rats, indicating that neurogenesis per se is not systematically associated with recovery from anhedonic-like behavior. Taken together, our data reveal for the first time an antidepressant action of gaboxadol and indicate a synergistic mechanism, regarding rapid onset of action and efficacy, when co-administered with escitalopram.


Subject(s)
Antidepressive Agents/therapeutic use , Citalopram/therapeutic use , Depression/drug therapy , GABA-A Receptor Agonists/therapeutic use , Isoxazoles/therapeutic use , Selective Serotonin Reuptake Inhibitors/therapeutic use , Animals , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Citalopram/pharmacology , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Depression/physiopathology , Disease Models, Animal , Drug Resistance , Drug Synergism , Drug Therapy, Combination , GABA-A Receptor Agonists/pharmacology , Isoxazoles/pharmacology , Male , Neurogenesis/drug effects , Neurons/drug effects , Raphe Nuclei/drug effects , Raphe Nuclei/physiopathology , Rats , Rats, Wistar , Selective Serotonin Reuptake Inhibitors/pharmacology , Synaptic Transmission/drug effects , Up-Regulation/drug effects
14.
Int J Neuropsychopharmacol ; 15(6): 715-25, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22008735

ABSTRACT

The aim of this proof-of-concept study was to compare the efficacy of escitalopram (20 mg/d) in combination with fixed doses of gaboxadol to escitalopram (20 mg) in the treatment of patients with severe major depressive disorder (MDD). Adult patients were randomized to 8 wk of double-blind treatment with fixed doses of placebo (n=71), escitalopram (20 mg, n=140), escitalopram (20 mg)+gaboxadol (5 mg) (n=139), or escitalopram (20 mg)+gaboxadol (10 mg) (n=140). The pre-defined primary analysis of efficacy was an analysis of covariance (ANCOVA) of change from baseline to endpoint (week 8) in Montgomery-Åsberg Depression Rating Scale (MADRS) total score using last observation carried forward (LOCF). There was no statistically significant difference in the mean change from baseline in MADRS total score between the 20 mg escitalopram+10 mg gaboxadol group and the 20 mg escitalopram group [difference=-0.45 MADRS points (95% CI -2.5 to 1.6, p=0.6619, full analysis set (FAS), LOCF, ANCOVA)] at week 8. The mean treatment differences to placebo at week 8 were -5.6 (95% CI -8.0 to -3.1, p<0.0001) (20 mg escitalopram), -5.1 (95% CI -7.5 to -2.7, p<0.0001) (20 mg escitalopram+5 mg gaboxadol), and -6.0 (95% CI -8.4 to -3.6, p<0.0001) (20 mg escitalopram+10 mg gaboxadol). The most common adverse events reported in the active treatment groups for which the incidence was higher than that in the placebo group, comprised nausea, anxiety and insomnia. There were no clinically relevant efficacy differences between a combination of escitalopram and gaboxadol compared to escitalopram alone in the treatment of severe MDD. All active treatment groups were superior in efficacy to placebo and were well tolerated.


Subject(s)
Analgesics/therapeutic use , Antidepressive Agents, Second-Generation/therapeutic use , Citalopram/therapeutic use , Depressive Disorder, Major/drug therapy , Isoxazoles/therapeutic use , Adolescent , Adult , Aged , Analysis of Variance , Double-Blind Method , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , International Cooperation , Male , Middle Aged , Psychiatric Status Rating Scales , Treatment Outcome , Young Adult
15.
J Pharmacol Exp Ther ; 338(1): 214-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21450931

ABSTRACT

Modulation of the extracellular levels of GABA via inhibition of the synaptic GABA transporter GAT1 by the clinically effective and selective GAT1 inhibitor tiagabine [(R)-N-[4,4-bis(3-methyl-2-thienyl)-3-butenyl]nipecotic acid; Gabitril] has proven to be an effective treatment strategy for focal seizures. Even though less is known about the therapeutic potential of other GABA transport inhibitors, previous investigations have demonstrated that N-[4,4-bis(3-methyl-2-thienyl)-3-butenyl]-3-hydroxy-4-(methylamino)-4,5,6,7-tetrahydrobenzo[d]isoxazol-3-ol (EF1502), which, like tiagabine, is inactive on GABA(A) receptors, inhibits both GAT1 and the extrasynaptic GABA and betaine transporter BGT1, and exerts a synergistic anticonvulsant effect when tested in combination with tiagabine. In the present study, the anticonvulsant activity and motor impairment associated with systemic administration of gaboxadol (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol), which, at the doses used in this study (i.e., 1-5 mg/kg) selectively activates extrasynaptic α4-containing GABA(A) receptors, was determined alone and in combination with either tiagabine or EF1502 using Frings audiogenic seizure-susceptible and CF1 mice. EF1502, when administered in combination with gaboxadol, resulted in reduced anticonvulsant efficacy and Rotarod impairment associated with gaboxadol. In contrast, tiagabine, when administered in combination with gaboxadol, did not modify the anticonvulsant action of gaboxadol or reverse its Rotarod impairment. Taken together, these results highlight the mechanistic differences between tiagabine and EF1502 and support a functional role for BGT1 and extrasynaptic GABA(A) receptors.


Subject(s)
Anticonvulsants/pharmacology , Ataxia/drug therapy , GABA Plasma Membrane Transport Proteins/physiology , Isoxazoles/pharmacology , Nipecotic Acids/pharmacology , Animals , Anticonvulsants/therapeutic use , Ataxia/physiopathology , Dose-Response Relationship, Drug , Female , GABA Agonists/pharmacology , Isoxazoles/therapeutic use , Male , Mice , Nipecotic Acids/therapeutic use , Receptors, GABA-A/physiology , Synapses/drug effects , Tiagabine
16.
Sleep ; 33(11): 1562-70, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21102998

ABSTRACT

OBJECTIVE: to evaluate cyclic alternating pattern (CAP) in a phase advance model of transient insomnia and the effects of gaboxadol and zolpidem. DESIGN: a randomized, double-blind, cross-over study in which habitual sleep time was advanced by 4 h. SETTING: 6 sleep research laboratories in US PARTICIPANTS: 55 healthy subjects (18-57 y) INTERVENTIONS: Gaboxadol 15 mg (GBX), zolpidem 10 mg (ZOL), and placebo (PBO). MEASUREMENTS: routine polysomnographic (PSG) measures, CAP, spectral power density, and self-reported sleep measures RESULTS: The phase advance model of transient insomnia produced significant changes in CAP parameters. Both GBX and ZOL significantly and differentially modified CAP parameters in the direction of more stable sleep. GBX brought the CAP rate in stage 1 sleep and slow wave sleep (SWS) closer to baseline levels but did not significantly change the CAP rate in stage 2. ZOL reduced the CAP rate in stage 2 to near baseline levels, whereas the CAP rate in stage 1 and SWS was reduced substantially below baseline levels. The CAP parameter A1 index (associated with SWS and sleep continuity) showed the highest correlation with self-reported sleep quality, higher than any traditional PSG, spectral, or other self-reported measures. CONCLUSION: disruptions in CAP produced by phase advanced sleep were significantly and differentially modulated by gaboxadol and zolpidem. The relative independence of CAP parameters from other electrophysiological measures of sleep, their high sensitivity to sleep disruption, and their strong association with subjective sleep quality suggest that CAP variables may serve as valuable endpoints in future insomnia research.


Subject(s)
GABA Agonists/pharmacology , GABA-A Receptor Agonists/pharmacology , Isoxazoles/pharmacology , Pyridines/pharmacology , Sleep Initiation and Maintenance Disorders/drug therapy , Sleep Stages/drug effects , Adolescent , Adult , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Middle Aged , Polysomnography/drug effects , Polysomnography/methods , Polysomnography/statistics & numerical data , Self Report , Young Adult , Zolpidem
17.
J Physiol ; 588(Pt 8): 1251-68, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20176630

ABSTRACT

The activation characteristics of synaptic and extrasynaptic GABA(A) receptors are important for shaping the profile of phasic and tonic inhibition in the central nervous system, which will critically impact on the activity of neuronal networks. Here, we study in isolation the activity of three agonists, GABA, muscimol and 4,5,6,7-tetrahydoisoxazolo[5,4-c]pyridin-3(2H)-one (THIP), to further understand the activation profiles of alpha 1 beta 3 gamma 2, alpha 4 beta 3 gamma 2 and alpha 4 beta 3 delta receptors that typify synaptic- and extrasynaptic-type receptors expressed in the hippocampus and thalamus. The agonists display an order of potency that is invariant between the three receptors, which is reliant mostly on the agonist dissociation constant. At delta subunit-containing extrasynaptic-type GABA(A) receptors, both THIP and muscimol additionally exhibited, to different degrees, superagonist behaviour. By comparing whole-cell and single channel currents induced by the agonists, we provide a molecular explanation for their different activation profiles. For THIP at high concentrations, the unusual superagonist behaviour on alpha 4 beta 3 delta receptors is a consequence of its ability to increase the duration of longer channel openings and their frequency, resulting in longer burst durations. By contrast, for muscimol, moderate superagonist behaviour was caused by reduced desensitisation of the extrasynaptic-type receptors. The ability to specifically increase the efficacy of receptor activation, by selected exogenous agonists over that obtained with the natural transmitter, may prove to be of therapeutic benefit under circumstances when synaptic inhibition is compromised or dysfunctional.


Subject(s)
Electrophysiological Phenomena/drug effects , GABA Agonists/pharmacology , Receptors, GABA-A/physiology , Synapses/physiology , Animals , Cell Line , Dose-Response Relationship, Drug , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Hydrogen-Ion Concentration , Isoxazoles/pharmacology , Kidney/cytology , Kidney/drug effects , Mice , Muscimol/pharmacology , Patch-Clamp Techniques , Receptors, GABA-A/drug effects , Synapses/drug effects , Transfection , gamma-Aminobutyric Acid/pharmacology
18.
Biochim Biophys Acta ; 1788(5): 1073-80, 2009 May.
Article in English | MEDLINE | ID: mdl-19366585

ABSTRACT

Changes in lipid bilayer elastic properties have been proposed to underlie the modulation of voltage-gated Na(+) and L-type Ca(2+) channels and GABA(A) receptors by amphiphiles. The amphiphile Triton X-100 increases the elasticity of lipid bilayers at micromolar concentrations, assessed from its effects on gramicidin channel A appearance rate and lifetime in artificial lipid bilayers. In the present study, the pharmacological action of Triton-X 100 on GABA(A) receptors expressed in Xenopus laevis oocytes was examined. Triton-X 100 inhibited GABA(A) alpha(1)beta(3)gamma(2S) receptor currents in a noncompetitive, time- and voltage-dependent manner and increased the apparent rate and extent of desensitization at 10 muM, which is 30 fold below the critical micelle concentration. In addition, Triton X-100 induced picrotoxin-sensitive GABA(A) receptor currents and suppressed allosteric modulation by flunitrazepam at alpha(1)beta(3)gamma(2S) receptors. All effects were independent of the presence of a gamma(2S) subunit in the GABA(A) receptor complex. The present study suggests that Triton X-100 may stabilize open and desensitized states of the GABA(A) receptor through changes in lipid bilayer elasticity.


Subject(s)
GABA Modulators/pharmacology , Octoxynol/pharmacology , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Animals , Biophysical Phenomena , Electrophysiological Phenomena , Female , Flunitrazepam/pharmacology , GABA-A Receptor Agonists , Humans , In Vitro Techniques , Lipid Bilayers , Oocytes/drug effects , Oocytes/metabolism , Picrotoxin/pharmacology , Recombinant Proteins/agonists , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Surface-Active Agents/pharmacology , Xenopus laevis
19.
Expert Opin Investig Drugs ; 18(2): 109-23, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19236259

ABSTRACT

The development of novel drugs falls into two completely different categories: truly novel drugs and drugs that can be considered as improvements of further advanced and eventually marketed drugs. The risk of failure and the reason for failure by these two classes of compounds obviously are very different. Truly novel drugs often rely on pharmacological data obtained in preclinical models paired with a scientific rationale for a mechanism thought to be relevant for the phenotype of the disease. The scientific insight into both the mechanisms underlying the disease and how these diseases can be manipulated by pharmacological means is therefore essential for the success of the drug. In practical terms, this means that a thorough understanding of the disease is a prerequisite for success. It is therefore a sobering thought that most of these compounds fail due to marginal efficacy in Phase II or III trials. The lack of success of these compounds may reflect either lack of knowledge of the disease, poor predictive value of the preclinical models, large heterogeneity in the underlying mechanisms for a given phenotype, or the use of the compound in a population that doesn't express a phenotype optimal for the drug. This year's list of discontinued compounds spans the range from truly innovative drugs to 'me-too' compounds and as such is highly useful in illustrating the current dilemmas for the pharmaceutical industry.


Subject(s)
Central Nervous System Agents , Peripheral Nervous System Agents , Pharmacology, Clinical/trends , Drug Industry , Humans , Mental Disorders/drug therapy
20.
J Neurosci Methods ; 178(2): 249-54, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19135084

ABSTRACT

The only method to quantify free extracellular levels of drugs in the brain of living animals is microdialysis. However, quantitative microdialysis has been hampered by methodological issues for decades. The problems arise from the need to establish the in vivo recovery for appropriate quantitation. In dealing with these issues the "dynamic no-net-flux" (DNNF) method seemed to be the experimental method of choice. Major disadvantages were, however, the need for a very high degree of bioanalytical precision and accuracy and the need for a large number of animals. Moreover, today we know that the experimental data are not always straightforward. To improve robustness and practicality of quantitative microdialysis sampling we modified the ultraslow microdialysis approach. Ultraslow microdialysis uses very low microdialysis flow rates (<200 nl/min) which increase recovery (both in vivo and in vitro) to over 90%. However, new practical issues arise when attempting to work with these flow rates. The resulting very low volumes and long lag times make this method very impractical for general application. In the modified version, addition of a carrier flow after the dialysis process has been completed, which negates the problems of long lag times and low volumes. The resulting dilution of the dialysis sample concentration can simply be mathematically corrected. In the current study we measured the free brain levels of two CNS compounds using the classic DNNF and the new modified ultraslow dialysis method. Modified ultraslow microdialysis was shown to generate robust data with the use of only small numbers of rats. The method is a promising tool for common straightforward screening of blood-brain barrier penetration of compounds into the brain.


Subject(s)
Brain/metabolism , Microdialysis/methods , Prefrontal Cortex/metabolism , Animals , Central Nervous System Agents/pharmacokinetics , Citalopram/pharmacokinetics , Extracellular Space/metabolism , Isoxazoles/pharmacokinetics , Male , Microdialysis/instrumentation , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...