Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 964
Filter
1.
Biochemistry (Mosc) ; 89(3): 507-522, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38648769

ABSTRACT

Some tricyclic antidepressants (TCAs), including amitriptyline (ATL), clomipramine (CLO), and desipramine (DES), are known to be effective for management of neuropathic pain. It was previously determined that ATL, CLO, and DES are capable of voltage-dependent blocking of NMDA receptors of glutamate (NMDAR), which play a key role in pathogenesis of neuropathic pain. Despite the similar structure of ATL, CLO, and DES, efficacy of their interaction with NMDAR varies significantly. In the study presented here, we applied molecular modeling methods to investigate the mechanism of binding of ATL, CLO, and DES to NMDAR and to identify structural features of the drugs that determine their inhibitory activity against NMDAR. Molecular docking of the studied TCAs into the NMDAR channel was performed. Conformational behavior of the obtained complexes in the lipid bilayer was simulated by the method of molecular dynamics (MD). A single binding site (upper) for the tertiary amines ATL and CLO and two binding sites (upper and lower) for the secondary amine DES were identified inside the NMDAR channel. The upper and lower binding sites are located along the channel axis at different distances from the extracellular side of the plasma membrane. MD simulation revealed that the position of DES in the lower site is stabilized only in the presence of sodium cation inside the NMDAR channel. DES binds more strongly to NMDAR compared to ATL and CLO due to simultaneous interaction of two hydrogen atoms of its cationic group with the asparagine residues of the ion pore of the receptor. This feature may be responsible for the stronger side effects of DES. It has been hypothesized that ATL binds to NMDAR less efficiently compared to DES and CLO due to its lower conformational mobility. The identified features of the structure- and cation-dependent mechanism of interaction between TCAs and NMDAR will help in the further development of effective and safe analgesic therapy.


Subject(s)
Antidepressive Agents, Tricyclic , Molecular Docking Simulation , Molecular Dynamics Simulation , Receptors, N-Methyl-D-Aspartate , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/chemistry , Antidepressive Agents, Tricyclic/pharmacology , Antidepressive Agents, Tricyclic/metabolism , Antidepressive Agents, Tricyclic/chemistry , Binding Sites , Amitriptyline/chemistry , Amitriptyline/metabolism , Amitriptyline/pharmacology , Humans , Clomipramine/pharmacology , Clomipramine/chemistry , Clomipramine/metabolism , Cations/metabolism , Cations/chemistry , Desipramine/pharmacology , Protein Binding
2.
Eur J Drug Metab Pharmacokinet ; 49(2): 181-190, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38172422

ABSTRACT

BACKGROUND AND OBJECTIVES: Doxepin, dosulepin, and clomipramine are tricyclic antidepressants (TCAs) that act as serotonin and noradrenaline reuptake inhibitors. The metabolites formed by N-dealkylation of these tricyclic antidepressants contribute to overall poor pharmacokinetics and efficacy. Deuteration of the methyl groups at metabolically active sites has been reported to be a useful strategy for developing more selective and potent antidepressants. This isotopic deuteration can lead to better bioavailability and overall effectiveness. The objective is to study the effect of site-selective deuteration of TCAs on their pharmacokinetic and pharmacodynamic profile by comparison with their nondeuterated counterparts. METHODS: In the current study, the pharmacokinetic profile and antidepressant behavior of deuterated TCAs were evaluated using the forced swim test (FST) and tail suspension test (TST), using male Wistar rats and male Swiss albino mice, respectively; additionally, a synaptosomal reuptake study was carried out. RESULTS: Compared with the nondeuterated parent drugs, deuterated forms showed improved efficacy in the behavior paradigm, indicating improved pharmacological activity. The pharmacokinetic parameters indicated increased maximum concentration in the plasma (Cmax), elimination half-life (t1/2), and area under the concentration-time curve (AUC)  in deuterated compounds. This can have a positive clinical impact on antidepressant treatment. Synaptosomal reuptake studies indicated marked inhibition of the reuptake mechanism of serotonin (5-HT) and norepinephrine. CONCLUSIONS: Deuterated TCAs can prove to be potentially better molecules in the treatment of neuropsychiatric disorders as compared with nondeuterated compounds. In addition, we have demonstrated a concept that metabolically active, site-selective deuteration can be beneficial for improving the pharmacokinetic and pharmacodynamic profiles of TCAs. A further toxicological study of these compounds is needed to validate their future clinical use.


Subject(s)
Antidepressive Agents, Tricyclic , Dothiepin , Rats , Mice , Animals , Male , Antidepressive Agents, Tricyclic/pharmacology , Antidepressive Agents, Tricyclic/therapeutic use , Clomipramine/pharmacology , Doxepin/pharmacology , Deuterium , Selective Serotonin Reuptake Inhibitors , Rats, Wistar , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Serotonin/metabolism , Models, Animal
3.
J Psychiatr Pract ; 29(6): 469-475, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37948171

ABSTRACT

Clomipramine (CIMI) is an effective treatment for obsessive-compulsive disorder in patients who have failed to respond to trials of selective serotonin transport inhibitors (eg, sertraline). The case presented here illustrates how knowledge of the pharmacodynamics and pharmacokinetics of CIMI in a specific patient can be used to personalize treatment to optimize the likelihood of efficacy (ie, maximum benefit to risk ratio). The approach described in this column considered: (1) the patient's diminished ability to clear CIMI and its major metabolite, desmethlyclomipramine due to a genetic deficiency in cytochrome P450 2D6 enzyme activity, and (2) the patient's ability to extensively convert CIMI to desmethlyclomipramine. That conversion impairs the ability to inhibit the serotonin transporter, the mechanism that is most likely responsible for the efficacy of CIMI in obsessive-compulsive disorder.


Subject(s)
Clomipramine , Obsessive-Compulsive Disorder , Humans , Clomipramine/pharmacology , Precision Medicine , Obsessive-Compulsive Disorder/drug therapy , Selective Serotonin Reuptake Inhibitors/pharmacology , Sertraline/pharmacology , Sertraline/therapeutic use
4.
Behav Brain Res ; 455: 114664, 2023 10 18.
Article in English | MEDLINE | ID: mdl-37714467

ABSTRACT

Depressive illness has been associated with impaired cognitive processes accompanied by reduced neurotrophin levels, especially brain-derived neurotrophic factor (BDNF), and dysfunctions in the hypothalamic-pituitary-adrenal (HPA) axis. In addition, depression is characterized by a decreased functioning of the serotonergic system due to changes in the activity or expression of its receptors including, most significantly, 5-HT1A, 5-HT2A, and 5-HT3 in brain regions that regulate mood, emotions, and memory, such as the prefrontal cortex, hippocampus, and amygdala. In this regard, rats treated with clomipramine (CMI) in the neonatal stage show depression-like behaviors that persist into adulthood; hence, this constitutes an adequate model of depression for exploring various molecular aspects associated with the etiology of this disorder. This, study, then, was designed to analyze the long-term effects of early postnatal exposure to CMI on the expression of 5-HT1A, 5-HT2A, and 5-HT3 receptors, as well as BDNF and GR in the following brain regions: PFC, amygdala, hippocampus, and hypothalamus, which could be related to alterations in memory and learning, as evaluated using the novel object recognition (NOR) and Morris water maze (MWM). Expression of the 5-HT1A, 5-HT2A, and 5-HT3 receptors, BDNF, and the glucocorticoid receptor (GR) was assessed by RT-qPCR in the four aforementioned brain regions, all of which play important roles in the control of memory and mood. Findings show that neonatal treatment with CMI causes alterations in memory and learning, as indicated by alterations in the results of the MWM and NOR tests. Expression of the 5-HT1A receptor increased in the hippocampus, amygdala, and hypothalamus, but decreased in the PFC, while the 5-HT2A and BDNF receptors decreased their expression in the PFC, amygdala, and hippocampus. There was no change in the expression of the 5-HT3 receptor. In addition, expression of GR in the hippocampus and PFC was low, but increased in the hypothalamus. Taken together, these data show that neonatal CMI treatment produces permanent molecular changes in brain regions related to learning and memory that could contribute to explaining the behavioral alterations observed in this model.


Subject(s)
Brain-Derived Neurotrophic Factor , Receptors, Glucocorticoid , Rats , Animals , Male , Receptors, Glucocorticoid/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Serotonin/metabolism , Clomipramine/pharmacology , Depression/metabolism , Brain/metabolism , Hippocampus/metabolism , Disease Models, Animal
5.
J Biochem ; 174(3): 267-272, 2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37137298

ABSTRACT

Three dynamin isoforms play critical roles in clathrin-dependent endocytosis. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) enters host cells via clathrin-dependent endocytosis. We previously reported that 3-(3-chloro-10,11-dihydro-5H-dibenzo[b,f]azepin-5-yl)-N,N-dimethylpropan-1-amine (clomipramine) inhibits the GTPase activity of dynamin 1, which is in mainly neuron. Therefore, we investigated whether clomipramine inhibits the activity of other dynamin isoforms in this study. We found that, similar to its inhibitory effect on dynamin 1, clomipramine inhibited the l-α-phosphatidyl-l-serine-stimulated GTPase activity of dynamin 2, which is expressed ubiquitously, and dynamin 3, which is expressed in the lung. Inhibition of GTPase activity raises the possibility that clomipramine can suppress SARS-CoV-2 entry into host cells.


Subject(s)
COVID-19 , Dynamin I , Humans , Clomipramine/pharmacology , Serine , Clathrin/pharmacology , SARS-CoV-2 , Dynamins , Endocytosis , Protein Isoforms
6.
Neurochem Res ; 47(11): 3464-3475, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35939172

ABSTRACT

Epigallocatechin 3-gallate (EGCG) is a natural polyphenolic antioxidant in green tea leaves with well-known health-promoting properties. However, the influence of EGCG on a chronic animal model of depression remains to be fully investigated, and the details of the molecular and cellular changes are still unclear. Therefore, the present study aimed to investigate the antidepressant effect of EGCG in mice subjected to chronic unpredictable mild stress (CUMS). After eight consecutive weeks of CUMS, the mice were treated with EGCG (200 mg/kg b.w.) by oral gavage for two weeks. A forced swimming test (FST) was used to assess depressive symptoms. EGCG administration significantly alleviated CUMS-induced depression-like behavior in mice. EGCG also effectively decreased serum interleukin-1ß (IL-1ß) and increased the mRNA expression levels of brain-derived neurotrophic factor (BDNF) in the hippocampal CA3 region of CUMS mice. Furthermore, electron microscopic examination of CA3 neurons in CUMS mice showed morphological features of apoptosis, loss or disruption of the myelin sheath, and degenerating synapses. These neuronal injuries were diminished with the administration of EGCG. The treatment effect of EGCG in CUMS-induced behavioral alterations was comparable with that of clomipramine hydrochloride (Anafranil), a tricyclic antidepressant drug. In conclusion, our study demonstrates that the antidepressive action of EGCG involves downregulation of serum IL-1ß, upregulation of BDNF mRNA in the hippocampus, and reduction of CA3 neuronal lesions.


Subject(s)
Brain-Derived Neurotrophic Factor , Depression , Interleukin-1beta , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Antidepressive Agents, Tricyclic/pharmacology , Antioxidants/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Catechin/analogs & derivatives , Clomipramine/pharmacology , Depression/drug therapy , Depression/etiology , Depression/metabolism , Disease Models, Animal , Hippocampus/metabolism , Interleukin-1beta/metabolism , Mice , RNA, Messenger/metabolism , Stress, Psychological/complications , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Tea/metabolism
7.
Transl Psychiatry ; 12(1): 245, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35688836

ABSTRACT

It is well known that neuroinflammation is closely related to the pathophysiology of depression. Due to individual differences in clinical research, the reduction of hippocampal volume in patients with depression is still controversial. In this experiment, we studied a typical kind of tricyclic antidepressant, clomipramine. We designed a series of experiments to find its role in depressive-like behavior, hippocampal neuroinflammation as well as hippocampal volume changes induced by chronic unpredictable mild stress (CMS). Rats exhibited defective behavior and hippocampal neuroinflammation after 12 weeks of CMS, which included elevated expression of cleaved interleukin-1ß (IL-1ß) and NLRP3 inflammasome together with the activation of microglia. Rats exposed to CMS showed weakened behavioral defects, reduced expression of IL-18, IL-6, and IL-1ß along with reversed activation of microglia after clomipramine treatment. This indicates that the antidepressant effect of clomipramine may be related to the reduced expression of NLRP3 inflammasome and cleaved IL-1ß. Moreover, we found an increased hippocampal volume in rats exposed to CMS after clomipramine treatment while CMS failed to affect hippocampal volume. All these results indicate that the NLRP3 inflammasome of microglia in the hippocampus is related to the antidepressant effects of clomipramine and CMS-induced depressive-like behavior in rats.


Subject(s)
Clomipramine , Inflammasomes , Animals , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacology , Clomipramine/metabolism , Clomipramine/pharmacology , Depression/drug therapy , Depression/metabolism , Disease Models, Animal , Hippocampus/metabolism , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats , Stress, Psychological/metabolism
8.
Int J Mol Sci ; 22(17)2021 Sep 04.
Article in English | MEDLINE | ID: mdl-34502505

ABSTRACT

BACKGROUND: Molecular mechanisms of depression remain unclear. The brain metabolome after antidepressant therapy is poorly understood and had not been performed for different routes of drug administration before the present study. Rats were exposed to chronic ultrasound stress and treated with intranasal and intraperitoneal clomipramine. We then analyzed 28 metabolites in the frontal cortex and hippocampus. METHODS: Rats' behavior was identified in such tests: social interaction, sucrose preference, forced swim, and Morris water maze. Metabolic analysis was performed with liquid chromatography. RESULTS: After ultrasound stress pronounced depressive-like behavior, clomipramine had an equally antidepressant effect after intranasal and intraperitoneal administration on behavior. Ultrasound stress contributed to changes of the metabolomic pathways associated with pathophysiology of depression. Clomipramine affected global metabolome in frontal cortex and hippocampus in a different way that depended on the route of administration. Intranasal route was associated with more significant changes of metabolites composition in the frontal cortex compared to the control and ultrasound groups while the intraperitoneal route corresponded with more profound changes in hippocampal metabolome compared to other groups. Since far metabolic processes in the brain can change in many ways depending on different routes of administration, the antidepressant therapy should also be evaluated from this point of view.


Subject(s)
Clomipramine/pharmacology , Depression/drug therapy , Administration, Intranasal/methods , Animals , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Clomipramine/administration & dosage , Depression/physiopathology , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Metabolome/physiology , Metabolomics/methods , Motor Activity/drug effects , Rats , Rats, Wistar , Stress, Psychological/drug therapy
9.
Invest Ophthalmol Vis Sci ; 62(7): 8, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34096973

ABSTRACT

Purpose: Patients that medicate with antidepressants commonly report dryness of eyes. The cause is often attributed to the anticholinergic properties of the drugs. However, regulation of tear production includes a substantial reflex-evoked component and is regulated via distinct centers in the brain. Further, the anticholinergic component varies greatly among antidepressants with different mechanisms of action. In the current study it was wondered if acute administration of antidepressants can disturb production of tears by affecting the afferent and/or central pathway. Methods: Tear production was examined in vivo in anesthetized rats in the presence or absence of the tricyclic antidepressant (TCA) clomipramine or the selective serotonin reuptake inhibitor (SSRI) escitalopram. The reflex-evoked production of tears was measured by challenging the surface of the eye with menthol (0.1 mM) and cholinergic regulation was examined by intravenous injection with the nonselective muscarinic agonist methacholine (1-5 µg/kg). Results: Acute administration of clomipramine significantly attenuated both reflex-evoked and methacholine-induced tear production. However, escitalopram only attenuated reflex-evoked tear production, while methacholine-induced production of tears remained unaffected. Conclusions: This study shows that antidepressants with different mechanisms of action can impair tear production by attenuating reflex-evoked signaling. Further, antimuscarinic actions are verified as a likely cause of lacrimal gland hyposecretion in regard to clomipramine but not escitalopram. Future studies on antidepressants with different selectivity profiles and mechanisms of action are required to further elucidate the mechanisms by which antidepressants affect tear production.


Subject(s)
Citalopram/pharmacology , Clomipramine/pharmacology , Dry Eye Syndromes , Evoked Potentials, Visual , Lacrimal Apparatus , Tears/physiology , Animals , Antidepressive Agents/pharmacology , Cholinergic Antagonists/pharmacology , Dry Eye Syndromes/chemically induced , Dry Eye Syndromes/physiopathology , Evoked Potentials, Visual/drug effects , Evoked Potentials, Visual/physiology , Lacrimal Apparatus/drug effects , Lacrimal Apparatus/physiology , Methacholine Chloride/pharmacology , Miotics/pharmacology , Rats
10.
Eur J Pharmacol ; 887: 173560, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32949600

ABSTRACT

We aimed at investigating the influence of clomipramine and selegiline administered in vivo in mice on lymphocyte subsets in lymphoid organs and SRBC-induced humoral immune response. Balb/c mice were given 7 or 14 oral doses (1 mg/kg) of selegiline or clomipramine. Lymphocyte B and T subsets and splenic regulatory T cell (Treg) subset were determined in non-immunized mice 24 and 72 h after the last dose of the drugs. Some mice treated with 7 doses were immunized with sheep red blood cells (SRBC) 2 h after the last dose, and their number of antibody forming cells, haemagglutinin titers and splenocyte subsets were determined. An increase in T lymphocytes and a decrease in B cells were visible in peripheral lymphoid organs, especially after 14 doses of selegiline or clomipramine in non-immunized mice, as well as in spleens of SRBC-immunized mice. The most pronounced change was a decrease in CD4+/CD8+ ratio resulting mainly from an increase in CD8+ subset after seven doses of the drugs in the non-immunized mice. However, it was of a transient nature, as it disappeared after 14 doses of the drugs. The tested drugs only slightly affected thymocyte maturation and did not alter Treg subset. Selegiline and clomipramine transiently stimulated IgG production in SRBC-immunized mice. Both selegiline and clomipramine administered in vivo modulated lymphocyte subsets. This immunomodulatory effect depended on the drug as well as duration of administration.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Clomipramine/pharmacology , Erythrocytes/drug effects , Immunity, Humoral/drug effects , Lymphocyte Subsets/drug effects , Monoamine Oxidase Inhibitors/pharmacology , Selegiline/pharmacology , T-Lymphocytes, Regulatory/drug effects , Animals , B-Lymphocytes/drug effects , CD4-CD8 Ratio , Female , Male , Mice , Mice, Inbred BALB C , Sheep , Spleen/cytology , Spleen/drug effects
11.
J Pharm Pharmacol ; 72(6): 836-842, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32144951

ABSTRACT

OBJECTIVES: Our aim was to find out whether clomipramine, a tricyclic antidepressant, and selegiline, a monoamine oxidase-B inhibitor, influence the activity of phagocytic cells after in-vivo administration in mice. METHODS: Clomipramine and selegiline were administered to Balb/c mice orally at a dose of 1 mg/kg, 7 or 14 times. IL-1ß and nitric oxide (NO) levels were measured in supernatants of the peritoneal macrophage cultures stimulated in vitro with lipopolysaccharide from Escherichia coli. The phagocytic activity of the granulocytes and monocytes was determined using a commercial Phagotest 24 and 72 h after the last dose of the investigated drugs. KEY FINDINGS: Seven doses of clomipramine or selegiline decreased IL-1ß production, while a rise in its synthesis was observed after 14 doses of selegiline. Clomipramine administered 14 times increased NO production. Clomipramine and selegiline administered seven times reduced the percentage of phagocytosing granulocytes. The drugs administered 14 times increased the percentage of phagocytosing granulocytes and decreased the percentage of phagocytosing monocytes. CONCLUSIONS: Both clomipramine and selegiline administered in vivo changed the phagocytic activity of blood cells and IL-1ß and NO production by murine peritoneal macrophages. This effect depended on the drug, the number of doses and the type of phagocytic cells.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Clomipramine/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Phagocytes/drug effects , Selegiline/pharmacology , Administration, Oral , Animals , Female , Granulocytes/drug effects , Interleukin-1beta/metabolism , Macrophages, Peritoneal/drug effects , Male , Mice , Mice, Inbred BALB C , Monocytes/drug effects , Nitric Oxide/metabolism , Phagocytosis/drug effects
12.
Eur J Pharmacol ; 873: 172963, 2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32007501

ABSTRACT

Preclinical and clinical studies have indicated that antidepressants can promote inflammation and fibrogenesis, particularly in the lung, by mechanisms not fully elucidated. We have previously shown that different classes of antidepressants can activate the lysophosphatidic acid (LPA) receptor LPA1, a major pathogenetic mediator of tissue fibrosis. The aim of the present study was to investigate whether in cultured human dermal and lung fibroblasts antidepressants could trigger LPA1-mediated profibrotic responses. In both cell types amitriptyline, clomipramine and mianserin mimicked the ability of LPA to induce the phosphorylation/activation of extracellular signal -regulated kinases 1 and 2 (ERK1/2), which was blocked by the selective LPA1 receptor antagonist AM966 and the LPA1/3 antagonist Ki16425. Antidepressant-induced ERK1/2 stimulation was absent in fibroblasts stably depleted of LPA1 by short hairpin RNA transfection and was prevented by pertussis toxin, an uncoupler of receptors from Gi/o proteins. Like LPA, antidepressants stimulated fibroblasts proliferation and this effect was blocked by either AM966 or the MEK1/2 inhibitor PD98059. Moreover, by acting through LPA1 antidepressants induced the expression of α-smooth muscle actin (α-SMA), a marker of myofibroblast differentiation, and caused an ERK1/2-dependent increase in the cellular levels of transforming growth factor-ß (TGF-ß)1, a potent fibrogenic cytokine. Pharmacological blockade of TGF-ß receptor type 1 prevented antidepressant- and LPA-induced α-SMA expression. These data indicate that in human dermal and lung fibroblasts different antidepressants can induce proliferative and differentiating responses by activating the LPA1 receptor coupled to ERK1/2 signalling and suggest that this property may contribute to the promotion of tissue fibrosis by these drugs.


Subject(s)
Antidepressive Agents/adverse effects , Fibrosis/chemically induced , Receptors, Lysophosphatidic Acid/drug effects , Actins/biosynthesis , Amitriptyline/pharmacology , Cell Proliferation/drug effects , Clomipramine/pharmacology , Cytokines/metabolism , Fibroblasts/drug effects , Fibrosis/pathology , Humans , MAP Kinase Signaling System/drug effects , Mianserin/pharmacology , RNA, Small Interfering/genetics , Receptor, Transforming Growth Factor-beta Type I/drug effects
13.
Eur J Med Chem ; 187: 111915, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31838329

ABSTRACT

A series of 10,11-dihydro-5H-dibenzo [b,f]azepine hydroxamates (4-15) were synthesized, behaving as histone deacetylase inhibitors, and examined for their influence on vascular cognitive impairment (VCI), which correlated with dementia. The results revealed that (E)-3-(4-(((3-(3-chloro-10,11-dihydro-5H-dibenzo [b,f]azepin-5-yl)propyl)amino)methyl)phenyl)-N-hydroxy-acrylamide (13) increases cerebral blood flow (CBF), attenuates cognitive impairment, and improves hippocampal atrophy in in vivo study. It is also able to increase the level of histone acetylation (H3K14 or H4K5) in the cortex and hippocampus of chronic cerebral hypoperfusion (CCH) mice; as a result, it could be a potential HDAC inhibitor for the treatment of vascular cognitive impairment.


Subject(s)
Azepines/pharmacology , Clomipramine/analogs & derivatives , Cognitive Dysfunction/drug therapy , Dementia, Vascular/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids/pharmacology , Protective Agents/pharmacology , Animals , Azepines/chemistry , Cell Line, Tumor , Clomipramine/chemistry , Clomipramine/pharmacology , Cognitive Dysfunction/metabolism , Dementia, Vascular/metabolism , Dose-Response Relationship, Drug , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Humans , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/chemistry , Male , Mice , Mice, Inbred C57BL , Molecular Structure , Protective Agents/chemical synthesis , Protective Agents/chemistry , Structure-Activity Relationship
14.
Int J Mol Sci ; 20(21)2019 Oct 29.
Article in English | MEDLINE | ID: mdl-31671916

ABSTRACT

We developed a pipeline for the discovery of transcriptomics-derived disease-modifying therapies and used it to validate treatments in vitro and in vivo that could be repurposed for TBI treatment. Desmethylclomipramine, ionomycin, sirolimus and trimipramine, identified by in silico LINCS analysis as candidate treatments modulating the TBI-induced transcriptomics networks, were tested in neuron-BV2 microglial co-cultures, using tumour necrosis factor α as a monitoring biomarker for neuroinflammation, nitrite for nitric oxide-mediated neurotoxicity and microtubule associated protein 2-based immunostaining for neuronal survival. Based on (a) therapeutic time window in silico, (b) blood-brain barrier penetration and water solubility, (c) anti-inflammatory and neuroprotective effects in vitro (p < 0.05) and (d) target engagement of Nrf2 target genes (p < 0.05), desmethylclomipramine was validated in a lateral fluid-percussion model of TBI in rats. Despite the favourable in silico and in vitro outcomes, in vivo assessment of clomipramine, which metabolizes to desmethylclomipramine, failed to demonstrate favourable effects on motor and memory tests. In fact, clomipramine treatment worsened the composite neuroscore (p < 0.05). Weight loss (p < 0.05) and prolonged upregulation of plasma cytokines (p < 0.05) may have contributed to the worsened somatomotor outcome. Our pipeline provides a rational stepwise procedure for evaluating favourable and unfavourable effects of systems-biology discovered compounds that modulate post-TBI transcriptomics.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Disease , Systems Biology/methods , Animals , Anti-Inflammatory Agents/pharmacology , Biomarkers , Cell Line , Clomipramine/analogs & derivatives , Clomipramine/metabolism , Clomipramine/pharmacology , Coculture Techniques , Cytokines/blood , Gene Expression , In Vitro Techniques , Ionomycin/pharmacology , Machine Learning , Male , Microglia/drug effects , Microglia/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotection , Neuroprotective Agents/pharmacology , Nitrites/metabolism , Rats , Sirolimus/pharmacology , Transcriptome , Trimipramine/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
15.
Brain Res ; 1724: 146443, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31513792

ABSTRACT

The participation of estrogens in depression has been well recognized. To exert its effects, estradiol binds mainly to estrogen receptors ESR1 and ESR2 (α and ß, respectively), expressed in brain regions including the hippocampus, limbic regions and hypothalamic nuclei. In rodents, modified estrogen receptors expression in brain areas have been implicated in different signs similar to those observed in depressive patients. Neonatal clomipramine (CMI) treatment is a pharmacological manipulation that generates behavioral and neurochemical changes that persist throughout adulthood and resemble human depression. The aim of this study was to analyze whether CMI neonatal treatment modifies the expression of nuclear ESR1 and ESR2 in the hippocampus, amygdala basolateral (BLA), amygdala medial (MeA), hypothalamic medial preoptic area (mPOA) and raphe nucleus in male rats. Our results indicate that CMI treatment significantly induced an mRNA increase of ESR1 in the hypothalamus, additionally produce a reduction in the mRNA ESR2 expression in raphe accompanied of an increase in hypothalamus and amygdala. CMI treated rats show more immunorreactive cells to ESR1 (ESR1-ir) in mPOA, BLA, MeA, together with a reduction of these cells in the hippocampal CA1 region. Moreover, an increase in the number of immunorreactive cells to ESR2 (ESR2-ir), in BLA and MeA, was observed in CMI treated rats. Additionally, the hippocampal CA2 region and raphe nucleus showed a decrease in these cells. Also, neonatal CMI treatment induced a decrease in the number of cells of the pyramidal layer in CA1. Overall, the results suggest that neonatal CMI treatment in rats (during brain development) induces changes in estrogen receptors in different brain areas involved with the regulation of depressive-like behaviors.


Subject(s)
Brain/metabolism , Clomipramine/pharmacology , Receptors, Estrogen/drug effects , Amygdala/metabolism , Animals , Animals, Newborn/metabolism , Behavior, Animal/drug effects , Clomipramine/metabolism , Depression/drug therapy , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Estrogens/metabolism , Hippocampus/metabolism , Hypothalamus/metabolism , Male , Raphe Nuclei/metabolism , Rats , Rats, Wistar , Receptors, Estrogen/metabolism , Sexual Behavior, Animal/drug effects
16.
Biomed Pharmacother ; 118: 109263, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31369988

ABSTRACT

It is well known that chamomile is one of the oldest known medicinal herbs and has been used to treat various disorders, but it is mainly German chamomile. The effects of Roman chamomile on depression still unclear. In this study, we used chronically stressed mice to investigate whether inhalation of Roman chamomile essential oil affects depression-like behavior. We previously reported that restraint and water immersion stress produce depression-like behavior and a blunted response to the tricyclic antidepressant clomipramine. Each mouse was exposed to restraint and water immersion stress for 15 days, and resistance to the effect of clomipramine was induced in a behavioral despair paradigm. In the present study, we found that cotreatment with clomipramine and inhalation of Roman chamomile attenuated depression-like behavior in a forced swim test. Next, we examined the hippocampal mRNA levels of two cytokines, tumor necrosis factor (TNF) alpha and interleukin-6 (IL-6); a neurotrophic factor, brain derived-neurotrophic factor (BDNF); and nerve growth factor (NGF). TNF alpha, IL-6 and BDNF mRNA levels did not change in the hippocampus of stressed mice. However, the NGF mRNA level was significantly decreased, and this decrease was not attenuated by treatment with clomipramine or inhalation of Roman chamomile alone. We also examined whether Roman chamomile combined with clomipramine treatment affects hippocampal neurogenesis and serum corticosterone levels. Stressed mice had fewer doublecortin (DCX)-positive cells in the subgranular zone of the dentate gyrus, but this was significantly attenuated by Roman chamomile and clomipramine treatment. In addition, the serum corticosterone level was also significantly decreased by treatment with Roman chamomile and clomipramine. These results suggest that Roman chamomile inhalation may enhance the antidepressant effect of clomipramine by increasing hippocampal neurogenesis and modulating corticosterone levels in patients with treatment-resistant depression.


Subject(s)
Behavior, Animal , Chamaemelum/chemistry , Clomipramine/therapeutic use , Depression/drug therapy , Inhalation Exposure , Plant Extracts/therapeutic use , Animals , Cell Proliferation/drug effects , Clomipramine/pharmacology , Corticosterone/blood , Cytokines/genetics , Cytokines/metabolism , Depression/blood , Doublecortin Protein , Drug Therapy, Combination , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Neurogenesis/drug effects , Plant Extracts/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Psychological/blood , Stress, Psychological/drug therapy
17.
Behav Brain Res ; 374: 112122, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31376442

ABSTRACT

Depression is a common mental disease affecting a lot of people of all ages around the world. Today, improving the therapeutic effects of currently used antidepressants such as clomipramine and, especially when they are administered at high doses is a topic of interest. The study aims to evaluate the eventual role of zinc (30 mg/Kg) in ameliorating clomipramine (75 mg/Kg) effects on behavior and oxidative stress equilibrium following a 6 day treatment in male Wistar rats. Our main findings showed that zinc improved clomipramine antidepressant and locomotor effects. Moreover, zinc reversed the oxidative stress induced by this drug in the liver. Thus, zinc at 30 mg/Kg may constitute an efficient adjuvant for clomipramine used at a high dose (75 mg/Kg) by boosting its efficacy on behavior and alleviating its negative effects on oxidative balance in liver.


Subject(s)
Locomotion/drug effects , Oxidative Stress/drug effects , Zinc/pharmacology , Animals , Antidepressive Agents/pharmacology , Clomipramine/pharmacology , Depression/drug therapy , Depressive Disorder/drug therapy , Disease Models, Animal , Male , Motor Activity/drug effects , Rats , Rats, Wistar , Stress, Psychological/drug therapy , Zinc/metabolism
18.
Int J Mol Sci ; 20(13)2019 Jul 09.
Article in English | MEDLINE | ID: mdl-31323957

ABSTRACT

Glioblastoma is the most common and malignant primary brain tumour in adults, with a dismal prognosis. This is partly due to considerable inter- and intra-tumour heterogeneity. Changes in the cellular energy-producing mitochondrial respiratory chain complex (MRC) activities are a hallmark of glioblastoma relative to the normal brain, and associate with differential survival outcomes. Targeting MRC complexes with drugs can also facilitate anti-glioblastoma activity. Whether mutations in the mitochondrial DNA (mtDNA) that encode several components of the MRC contribute to these phenomena remains underexplored. We identified a germ-line mtDNA mutation (m. 14798T > C), enriched in glioblastoma relative to healthy controls, that causes an amino acid substitution F18L within the core mtDNA-encoded cytochrome b subunit of MRC complex III. F18L is predicted to alter corresponding complex III activity, and sensitivity to complex III-targeting drugs. This could in turn alter reactive oxygen species (ROS) production, cell behaviour and, consequently, patient outcomes. Here we show that, despite a heterogeneous mitochondrial background in adult glioblastoma patient biopsy-derived cell cultures, the F18L substitution associates with alterations in individual MRC complex activities, in particular a 75% increase in MRC complex II_III activity, and a 34% reduction in CoQ10, the natural substrate for MRC complex III, levels. Downstream characterisation of an F18L-carrier revealed an 87% increase in intra-cellular ROS, an altered cellular distribution of mitochondrial-specific ROS, and a 64% increased sensitivity to clomipramine, a repurposed MRC complex III-targeting drug. In patients, F18L-carriers that received the current standard of care treatment had a poorer prognosis than non-carriers (373 days vs. 415 days, respectively). Single germ-line mitochondrial mutations could predispose individuals to differential prognoses, and sensitivity to mitochondrial targeted drugs. Thus, F18L, which is present in blood could serve as a useful non-invasive biomarker for the stratification of patients into prognostically relevant groups, one of which requires a lower dose of clomipramine to achieve clinical effect, thus minimising side-effects.


Subject(s)
DNA, Mitochondrial/genetics , Germ-Line Mutation/genetics , Glioblastoma/genetics , Clomipramine/pharmacology , Humans , Kaplan-Meier Estimate , Male , Mitochondria/metabolism , Mutation/genetics , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/metabolism
19.
J Affect Disord ; 254: 15-25, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31082627

ABSTRACT

BACKGROUND: Depression has recently been referred to as a neuroimmune disease because it is characterized by inflammatory changes in the cerebral cortex and hippocampus. Studies have demonstrated that microglial activation plays a crucial role in releasing inflammatory cytokines in the central nervous system (CNS), thereby contributing to depression, the mechanism underlying which remains unclear. METHODS: First, we examined microglial activation and inflammatory changes in C57BL/6 male mice injected with lipopolysaccharide (LPS; 1 mg/kg), which leads to depressive behaviors in mice that were attenuated by the antidepressant clomipramine. Second, we utilized a BV2 cell line and primary microglial cultures to determine the inflammatory response in vitro, and the effects of clomipramine exerted on the inflammatory response using real-time polymerase chain reaction and ELISA. Third, we utilized NLRP3 (NOD-like receptor protein 3) knock-out (KO) mice to prove that NLRP3 is involved in the effects of clomipramine. RESULTS: The results showed that LPS injection induced depressive-like behaviors in mice, as assessed using several behavioral tests including body weight, and forced swimming and tail suspension tests. The LPS-induced expression of interleukin-1beta (IL-1ß), IL-6, and tumor necrosis factor alpha could be downregulated by clomipramine pre-treatment both in vivo and in vitro. The inhibitory effect of clomipramine on the LPS-induced increase in cytokines was found at both the protein and gene levels. Clomipramine significantly reduced the LPS-induced increase in NLRP3 gene expression in BV2 cells. Furthermore, we utilized NLRP3 KO mice to explore whether NLPP3 was involved in this process and found that clomipramine treatment inhibits the LPS-induced increased expression of IL-1ß. CONCLUSION: These results imply that clomipramine could attenuate depressive behaviors and neuroinflammation induced by LPS via partial regulation of NLRP3.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antidepressive Agents/pharmacology , Clomipramine/pharmacology , Microglia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Cytokines/metabolism , Depression/chemically induced , Depression/drug therapy , Depressive Disorder/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Inflammasomes/drug effects , Inflammasomes/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
20.
Sci Rep ; 9(1): 4881, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30890728

ABSTRACT

Antidepressants are commonly prescribed psychotropic substances for the symptomatic treatment of mood disorders. Their primary mechanism of action is the modulation of neurotransmission and the consequent accumulation of monoamines, such as serotonin and noradrenaline. However, antidepressants have additional molecular targets that, through multiple signaling cascades, may ultimately alter essential cellular processes. In this regard, it was previously demonstrated that clomipramine, a widely used FDA-approved tricyclic antidepressant, interferes with the autophagic flux and severely compromises the viability of tumorigenic cells upon cytotoxic stress. Consistent with this line of evidence, we report here that clomipramine undermines autophagosome formation and cargo degradation in primary dissociated neurons. A similar pattern was observed in the frontal cortex and liver of treated mice, as well as in the nematode Caenorhabditis elegans exposed to clomipramine. Together, our findings indicate that clomipramine may negatively regulate the autophagic flux in various tissues, with potential metabolic and functional implications for the homeostatic maintenance of differentiated cells.


Subject(s)
Affective Disorders, Psychotic/drug therapy , Antidepressive Agents, Tricyclic/pharmacology , Clomipramine/pharmacology , Neurons/drug effects , Affective Disorders, Psychotic/pathology , Animals , Antidepressive Agents, Tricyclic/adverse effects , Autophagy/drug effects , Caenorhabditis elegans/drug effects , Clomipramine/adverse effects , Disease Models, Animal , Liver/drug effects , Liver/metabolism , Mice , Neurons/metabolism , Norepinephrine/metabolism , Serotonin/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...