Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.185
Filter
Add more filters

Publication year range
1.
Mol Cell Neurosci ; 125: 103842, 2023 06.
Article in English | MEDLINE | ID: mdl-36924917

ABSTRACT

Chemical platforms that facilitate both the identification and elucidation of new areas for therapeutic development are necessary but lacking. Activity-based protein profiling (ABPP) leverages active site-directed chemical probes as target discovery tools that resolve activity from expression and immediately marry the targets identified with lead compounds for drug design. However, this approach has traditionally focused on predictable and intrinsic enzyme functionality. Here, we applied our activity-based proteomics discovery platform to map non-encoded and post-translationally acquired enzyme functionalities (e.g. cofactors) in vivo using chemical probes that exploit the nucleophilic hydrazine pharmacophores found in a classic antidepressant drug (e.g. phenelzine, Nardil®). We show the probes are in vivo active and can map proteome-wide tissue-specific target engagement of the drug. In addition to engaging targets (flavoenzymes monoamine oxidase A/B) that are associated with the known therapeutic mechanism as well as several other members of the flavoenzyme family, the probes captured the previously discovered N-terminal glyoxylyl (Glox) group of Secernin-3 (SCRN3) in vivo through a divergent mechanism, indicating this functional feature has biochemical activity in the brain. SCRN3 protein is ubiquitously expressed in the brain, yet gene expression is regulated by inflammatory stimuli. In an inflammatory pain mouse model, behavioral assessment of nociception showed Scrn3 male knockout mice selectively exhibited impaired thermal nociceptive sensitivity. Our study provides a guided workflow to entangle molecular (off)targets and pharmacological mechanisms for therapeutic development.


Subject(s)
Nociception , Phenelzine , Animals , Mice , Male , Phenelzine/pharmacology , Proteome , Nerve Tissue Proteins
2.
Drug Chem Toxicol ; 47(1): 81-89, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37246945

ABSTRACT

Acetaminophen (APAP) overdosing is the most common cause of drug-induced liver failure. Despite extensive study, N-acetylcysteine is currently the only antidote utilized for treatment. The purpose of this study was to evaluate the effect and mechanisms of phenelzine, an FDA-approved antidepressant, on APAP-induced toxicity in HepG2 cells. The human liver hepatocellular cell line HepG2 was used to investigate APAP-induced cytotoxicity. The protective effects of phenelzine were determined by examining the cell viability, combination index calculation, Caspase 3/7 activation, Cytochrome c release, H2O2 levels, NO levels, GSH activity, PERK protein levels, and pathway enrichment analysis. Elevated H2O2 production and decreased glutathione (GSH) levels were indicators of APAP-induced oxidative stress. The combination index of 2.04 indicated that phenelzine had an antagonistic effect on APAP-induced toxicity. When compared to APAP alone, phenelzine treatment considerably reduced caspase 3/7 activation, cytochrome c release, and H2O2 generation. However, phenelzine had minimal effect on NO and GSH levels and did not alleviate ER stress. Pathway enrichment analysis revealed a potential connection between APAP toxicity and phenelzine metabolism. These findings suggested that phenelzine's protective effect against APAP-induced cytotoxicity could be attributed to the drug's capacity to reduce APAP-mediated apoptotic signaling.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Humans , Acetaminophen/toxicity , Hep G2 Cells , Phenelzine/metabolism , Phenelzine/pharmacology , Caspase 3/metabolism , Cytochromes c/metabolism , Hydrogen Peroxide/pharmacology , Liver , Oxidative Stress , Apoptosis , Chemical and Drug Induced Liver Injury/metabolism , Glutathione/metabolism
3.
J Clin Psychopharmacol ; 43(2): 149-151, 2023.
Article in English | MEDLINE | ID: mdl-36700727

ABSTRACT

PURPOSE/BACKGROUND: Despite several decades of research, there are no US Food and Drug Administration-approved medications for trichotillomania or medications generally approved in other geographical jurisdictions. Monoamine oxidase inhibitors show efficacy in the treatment of depression and some possible promise for obsessive compulsive disorder. METHODS/PROCEDURES: We present new data from a case series collected in a specialty clinical practice over a 4-year period. FINDINGS/RESULTS: In 5 treatment-resistant patients whose trichotillomania had not improved with at least 1 course of cognitive behavior therapy and trials of n -acetyl cysteine, an antipsychotic, and a serotonin selective reuptake inhibitor, 2 had marked clinical improvement (>40% improvement) on phenelzine, 1 improved on tranylcypromine, and 2 showed no improvement (<10%) on phenelzine. In 2 of the 3 patients who experienced improvement, there was co-occurring depression. IMPLICATIONS/CONCLUSIONS: Monoamine oxidase inhibitors in trichotillomania may deserve large-scale randomized controlled trials, particularly in specialist settings where first-line interventions have proven inadequate to manage severe symptoms.


Subject(s)
Obsessive-Compulsive Disorder , Trichotillomania , United States , Humans , Monoamine Oxidase Inhibitors/therapeutic use , Trichotillomania/drug therapy , Phenelzine , Obsessive-Compulsive Disorder/drug therapy , Selective Serotonin Reuptake Inhibitors
4.
Cell Mol Neurobiol ; 42(1): 225-242, 2022 Jan.
Article in English | MEDLINE | ID: mdl-33839994

ABSTRACT

Phenelzine (PLZ) is a monoamine oxidase (MAO)-inhibiting antidepressant with anxiolytic properties. This multifaceted drug has a number of pharmacological and neurochemical effects in addition to inhibition of MAO, and findings on these effects have contributed to a body of evidence indicating that PLZ also has neuroprotective/neurorescue properties. These attributes are reviewed in this paper and include catabolism to the active metabolite ß-phenylethylidenehydrazine (PEH) and effects of PLZ and PEH on the GABA-glutamate balance in brain, sequestration of reactive aldehydes, and inhibition of primary amine oxidase. Also discussed are the encouraging findings of the effects of PLZ in animal models of stroke, spinal cord injury, traumatic brain injury, and multiple sclerosis, as well other actions such as reduction of nitrative stress, reduction of the effects of a toxin on dopaminergic neurons, potential anticonvulsant actions, and effects on brain-derived neurotrophic factor, neural cell adhesion molecules, an anti-apoptotic factor, and brain levels of ornithine and N-acetylamino acids.


Subject(s)
Antidepressive Agents , Monoamine Oxidase Inhibitors , Neuroprotective Agents , Phenelzine , Animals , Antidepressive Agents/pharmacology , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Neuroprotective Agents/pharmacology , Phenelzine/pharmacology , Rats , Rats, Sprague-Dawley
5.
J Clin Psychopharmacol ; 41(4): 461-464, 2021.
Article in English | MEDLINE | ID: mdl-34108430

ABSTRACT

PURPOSE/BACKGROUND: Despite the availability of a range of efficacious evidence-based treatments for obsessive-compulsive disorder (OCD), not all patients experience sufficient benefit or are able to tolerate them in practice. Monoamine oxidase inhibitors (MAOIs) show efficacy in the treatment of depression and certain anxiety disorders (such as social anxiety disorder). METHODS/PROCEDURES: We survey the evidence base from case reports, and clinical trials, regarding use of MAOIs in OCD. We then present new data from a case series collected in routine clinical practice in a specialist clinical service. FINDINGS/RESULTS: In 9 treatment-resistant patients whose OCD had not improved with at least 2 standard treatment trials, 3 had marked clinical improvement (>35% improvement on YBOCS) on phenelzine, 3 had some improvement (15-34.9%), and 3 showed minimal or no improvement (<15%). In the 3 patients who experienced minimal/no improvement, 2 had discontinued early because of lack of tolerability, and the other patient discontinued after 4 weeks because of perceived lack of symptom benefit. IMPLICATIONS/CONCLUSIONS: We suggest that (1) MAOIs in treatment-resistant OCD require appropriate research scrutiny in large-scale randomized controlled trials; and (2) MAOIs merit consideration as a treatment option in individual cases of OCD, particularly in specialist settings where first-line interventions have proven inadequate to manage severe symptoms.


Subject(s)
Behavioral Symptoms , Obsessive-Compulsive Disorder , Phenelzine , Adult , Behavioral Symptoms/diagnosis , Behavioral Symptoms/drug therapy , Clinical Trials as Topic , Drug Resistance , Female , Humans , Male , Monoamine Oxidase Inhibitors/administration & dosage , Monoamine Oxidase Inhibitors/adverse effects , Obsessive-Compulsive Disorder/diagnosis , Obsessive-Compulsive Disorder/drug therapy , Obsessive-Compulsive Disorder/psychology , Phenelzine/administration & dosage , Phenelzine/adverse effects , Risk Assessment , Treatment Outcome
6.
Prostate ; 79(6): 667-677, 2019 05.
Article in English | MEDLINE | ID: mdl-30693539

ABSTRACT

BACKGROUND: Monoamine oxidase A (MAOA) is best known for its role in neuro-transmitter regulation. Monoamine oxidase inhibitors are used to treat atypical depression. MAOA is highly expressed in high grade prostate cancer and modulates tumorigenesis and progression in prostate cancer. Here, we investigated the potential role of MAOA inhibitors (MAOAIs) in relation to the androgen receptor (AR) pathway and resistance to antiandrogen treatment in prostate cancer. METHODS: We examined MAOA expression and the effect of MAOI treatment in relation to AR-targeted treatments using the LNCaP, C4-2B, and 22Rv1 human prostate cancer cell lines. MAOA, AR-full length (AR-FL), AR splice variant 7 (AR-V7), and PSA expression was evaluated in the presence of MAOAIs (clorgyline, phenelzine), androgenic ligand (R1881), and antiandrogen (enzalutamide) treatments. An enzalutamide resistance cell line was generated to test the effect of MAOAI treatment in this model. RESULTS: We observed that MAOAIs, particularly clorgyline and phenelzine, were effective at decreasing MAOA activity in human prostate cancer cells. MAOAIs significantly decreased growth of LNCaP, C4-2B, and 22Rv1 cells and produced additive growth inhibitory effects when combined with enzalutamide. Clorgyline decreased expression of AR-FL and AR-V7 in 22Rv1 cells and was effective at decreasing growth of an enzalutamide-resistant C4-2B cell line with increased AR-V7 expression. CONCLUSIONS: MAOAIs decrease growth and proliferation of androgen-sensitive and castration-resistant prostate cancer cells. Clorgyline, in particular, decreases expression of AR-FL and AR-V7 expression and decreases growth of an enzalutamide-resistant cell line. These findings provide preclinical validation of MAOA inhibitors either alone or in combination with antiandrogens for therapeutic intent in patients with advanced forms of prostate cancer.


Subject(s)
Clorgyline/pharmacology , Drug Resistance, Neoplasm/drug effects , Phenelzine/pharmacology , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms, Castration-Resistant , Receptors, Androgen/metabolism , Androgen Antagonists/pharmacology , Benzamides , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/pharmacology , Neoplasm Grading , Nitriles , Phenylthiohydantoin/pharmacology , Prostate/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology
7.
J Pharmacol Exp Ther ; 371(2): 555-566, 2019 11.
Article in English | MEDLINE | ID: mdl-31270215

ABSTRACT

Novel mechanisms and health benefits have been recently suggested for the antidepressant drug phenelzine (PHE), known as a nonselective monoamine oxidase inhibitor. They include an antilipogenic action that could have an impact on excessive fat accumulation and obesity-related metabolic alterations. We evaluated the metabolic effects of an oral PHE treatment on mice fed a high-fat diet (HFD). Eleven-week-old male C57BL/6 mice were fed a HFD and either a 0.028% PHE solution (HFD + PHE) or water to drink for 11 weeks. PHE attenuated the increase in body weight and adiposity without affecting food consumption. Energy efficiency was lower in HFD + PHE mice. Lipid content was reduced in subcutaneous fat pads, liver, and skeletal muscle. In white adipose tissue (WAT), PHE reduced sterol regulatory element-binding protein-1c and phosphoenolpyruvate carboxykinase mRNA levels, inhibited amine-induced lipogenesis, and did not increase lipolysis. Moreover, HFD + PHE mice presented diminished levels of hydrogen peroxide release in subcutaneous WAT and reduced expression of leukocyte transmigration markers and proinflammatory cytokines in visceral WAT and liver. PHE reduced the circulating levels of glycerol, triacylglycerols, high-density lipoprotein cholesterol, and insulin. Insulin resistance was reduced, without affecting glucose levels and glucose tolerance. In contrast, PHE increased rectal temperature and slightly increased energy expenditure. The mitigation of HFD-induced metabolic disturbances points toward a promising role for PHE in obesity treatment and encourages further research on its mechanisms of action. SIGNIFICANCE STATEMENT: Phenelzine reduces body fat, markers of oxidative stress, inflammation, and insulin resistance in high-fat diet mice. Semicarbazide-sensitive amine oxidase, monoamine oxidase, phosphoenolpyruvate carboxykinase, and sterol regulatory element-binding protein-1c are involved in the metabolic effects of phenelzine. Phenelzine could be potentially used for the treatment of obesity-related complications.


Subject(s)
Adipose Tissue/drug effects , Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Insulin Resistance/physiology , Monoamine Oxidase Inhibitors/administration & dosage , Phenelzine/administration & dosage , Administration, Oral , Animals , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Oxidative Stress/physiology , Treatment Outcome
8.
Drug Metab Dispos ; 47(4): 436-443, 2019 04.
Article in English | MEDLINE | ID: mdl-30709838

ABSTRACT

Clopidogrel acyl-ß-d-glucuronide is a mechanism-based inhibitor of cytochrome P450 2C8 in human liver microsomes (HLMs). However, time-dependent inactivation (TDI) of CYP2C8 could not be detected in an earlier study in human recombinant CYP2C8 (Supersomes). Here, we investigate whether different enzyme sources exhibit differences in detection of CYP2C8 TDI under identical experimental conditions. Inactivation of CYP2C8 by amiodarone (100 µM), clopidogrel acyl-ß-d-glucuronide (100 µM), gemfibrozil 1-O-ß-glucuronide (100 µM), and phenelzine (100 µM) was investigated in HLMs and three recombinant human CYP2C8 preparations (Supersomes, Bactosomes, and EasyCYP Bactosomes) using amodiaquine N-deethylation as the marker reaction. Furthermore, the inactivation kinetics of CYP2C8 by clopidogrel glucuronide (5-250 µM) was determined in Supersomes and Bactosomes. Amiodarone caused weak TDI in all enzyme preparations tested, while the extent of inactivation by clopidogrel glucuronide, gemfibrozil glucuronide, and phenelzine varied markedly between preparations, and even different Supersome lots. Both glucuronides caused strong inactivation of CYP2C8 in HLMs, Bactosomes and in one Supersome lot (>50% inhibition), but significant inactivation could not be reliably detected in other Supersome lots or EasyCYP Bactosomes. In Bactosomes, the concentration producing half of kinact (KI) and maximal inactivation rate (kinact) of clopidogrel glucuronide (14 µM and 0.054 minute-1) were similar to those determined previously in HLMs. Phenelzine caused strong inactivation of CYP2C8 in one Supersome lot (91% inhibition) but not in HLMs or other recombinant CYP2C8 preparations. In conclusion, different enzyme sources and different lots of the same recombinant enzyme preparation are not equally sensitive to detect inactivation of CYP2C8, suggesting that recombinant CYPs should be avoided when identifying mechanism-based inhibitors.


Subject(s)
Cytochrome P-450 CYP2C8/metabolism , Amiodarone/metabolism , Clopidogrel/metabolism , Gemfibrozil/metabolism , Glucuronides/metabolism , Humans , Kinetics , Microsomes, Liver/metabolism , Phenelzine/metabolism , Sensitivity and Specificity
9.
Molecules ; 24(21)2019 Oct 31.
Article in English | MEDLINE | ID: mdl-31683720

ABSTRACT

Drug-drug interactions (DDIs) severity assessment is a crucial problem because polypharmacy is increasingly common in modern medical practice. Many DDIs are caused by alterations of the plasma concentrations of one drug due to another drug inhibiting and/or inducing the metabolism or transporter-mediated disposition of the victim drug. Accurate assessment of clinically relevant DDIs for novel drug candidates represents one of the significant tasks of contemporary drug research and development and is important for practicing physicians. This work is a development of our previous investigations and aimed to create a model for the severity of DDIs prediction. PASS program and PoSMNA descriptors were implemented for prediction of all five classes of DDIs severity according to OpeRational ClassificAtion (ORCA) system: contraindicated (class 1), provisionally contraindicated (class 2), conditional (class 3), minimal risk (class 4), no interaction (class 5). Prediction can be carried out both for known drugs and for new, not yet synthesized substances using only their structural formulas. Created model provides an assessment of DDIs severity by prediction of different ORCA classes from the first most dangerous class to the fifth class when DDIs do not take place in the human organism. The average accuracy of DDIs class prediction is about 0.75.


Subject(s)
Drug Interactions , Enzyme Inhibitors/pharmacology , Enzyme Activation/drug effects , Phenelzine/chemistry , Tranylcypromine/chemistry
10.
J Neurovirol ; 24(2): 192-203, 2018 04.
Article in English | MEDLINE | ID: mdl-29256041

ABSTRACT

Most studies of HIV latency focus on the peripheral population of resting memory T cells, but the brain also contains a distinct reservoir of HIV-infected cells in microglia, perivascular macrophages, and astrocytes. Studying HIV in the brain has been challenging, since live cells are difficult to recover from autopsy samples and primate models of SIV infection utilize viruses that are more myeloid-tropic than HIV due to the expression of Vpx. Development of a realistic small animal model would greatly advance studies of this important reservoir and permit definitive studies of HIV latency. When radiation or busulfan-conditioned, immune-deficient NSG mice are transplanted with human hematopoietic stem cells, human cells from the bone marrow enter the brain and differentiate to express microglia-specific markers. After infection with replication competent HIV, virus was detected in these bone marrow-derived human microglia. Studies of HIV latency in this model would be greatly enhanced by the development of compounds that can selectively reverse HIV latency in microglial cells. Our studies have identified members of the CoREST repression complex as key regulators of HIV latency in microglia in both rat and human microglial cell lines. The monoamine oxidase (MAO) and potential CoREST inhibitor, phenelzine, which is brain penetrant, was able to stimulate HIV production in human microglial cell lines and human glial cells recovered from the brains of HIV-infected humanized mice. The humanized mice we have developed therefore show great promise as a model system for the development of strategies aimed at defining and reducing the CNS reservoir.


Subject(s)
AIDS Dementia Complex/drug therapy , Anti-HIV Agents/pharmacology , HIV-1/drug effects , Microglia/drug effects , Monoamine Oxidase Inhibitors/pharmacology , Nerve Tissue Proteins/genetics , Phenelzine/pharmacology , Repressor Proteins/genetics , AIDS Dementia Complex/genetics , AIDS Dementia Complex/physiopathology , AIDS Dementia Complex/virology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/virology , Brain/drug effects , Brain/physiopathology , Brain/virology , Busulfan/toxicity , Cell Differentiation , Co-Repressor Proteins , Disease Models, Animal , Gene Expression Regulation , HIV-1/pathogenicity , HIV-1/physiology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/virology , Nerve Tissue Proteins/metabolism , Rats , Repressor Proteins/metabolism , Transplantation, Heterologous , Virus Latency/drug effects , Virus Latency/genetics , Whole-Body Irradiation
11.
Int J Mol Sci ; 19(10)2018 Sep 25.
Article in English | MEDLINE | ID: mdl-30257452

ABSTRACT

Phenelzine has been suggested to have an antiobesity effect by inhibiting de novo lipogenesis, which led us to investigate the metabolic effects of oral chronic phenelzine treatment in high-sucrose-drinking mice. Sucrose-drinking mice presented higher body weight gain and adiposity versus controls. Phenelzine addition did not decrease such parameters, even though fat pad lipid content and weights were not different from controls. In visceral adipocytes, phenelzine did not impair insulin-stimulated de novo lipogenesis and had no effect on lipolysis. However, phenelzine reduced the mRNA levels of glucose transporters 1 and 4 and phosphoenolpyruvate carboxykinase in inguinal white adipose tissue (iWAT), and altered circulating levels of free fatty acids (FFA) and glycerol. Interestingly, glycemia was restored in phenelzine-treated mice, which also had higher insulinaemia. Phenelzine-treated mice presented higher rectal temperature, which was associated to reduced mRNA levels of uncoupling protein 1 in brown adipose tissue. Furthermore, unlike sucrose-drinking mice, hepatic malondialdehyde levels were not altered. In conclusion, although de novo lipogenesis was not inhibited by phenelzine, the data suggest that the ability to re-esterify FFA is impaired in iWAT. Moreover, the effects on glucose homeostasis and oxidative stress suggest that phenelzine could alleviate obesity-related alterations and deserves further investigation in obesity models.


Subject(s)
Monoamine Oxidase Inhibitors/therapeutic use , Obesity/drug therapy , Obesity/etiology , Phenelzine/therapeutic use , Sucrose/adverse effects , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adiposity/drug effects , Administration, Oral , Animals , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/metabolism , Lipogenesis/drug effects , Male , Mice , Mice, Inbred C57BL , Monoamine Oxidase Inhibitors/administration & dosage , Obesity/blood , Obesity/metabolism , Oxidative Stress/drug effects , Phenelzine/administration & dosage , Sucrose/metabolism , Weight Gain/drug effects
12.
CNS Spectr ; 22(5): 391-396, 2017 10.
Article in English | MEDLINE | ID: mdl-28004621

ABSTRACT

Irreversible monoamine oxidase inhibitor (MAOI) antidepressants have significant efficacy in treatment-resistant unipolar depression, but in some instances patients may not achieve remission. Among the adjunctive and augmentation strategies, certain second-generation antipsychotics (SGAs) have approval for inadequate responders to antidepressant therapy, including aripiprazole, brexpiprazole, and quetiapine, with lurasidone and the olanzapine/fluoxetine combination indicated for bipolar depression. Clinicians may eschew SGA options in part due to the limited literature on SGA-MAOI combinations, with only one published case involving aripiprazole, and none for olanzapine, lurasidone, or brexpiprazole. In addition to the limited publication history on SGA-MAOI treatment, clinicians may also be deterred by uncertainty regarding SGA mechanisms and the risk of serotonin syndrome or other adverse outcomes. This paper describes the case of a 54-year-old male with a history of psychotic unipolar depression treated with a combination of phenelzine, aripiprazole, and quetiapine, and reviews the 12 published cases of SGA-MAOI combination therapy with a focus on the pharmacological basis for serotonin syndrome, and the SGA mechanisms that should not be associated with a risk for this syndrome.


Subject(s)
Affective Disorders, Psychotic/drug therapy , Antidepressive Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Monoamine Oxidase Inhibitors/therapeutic use , Antidepressive Agents/administration & dosage , Antipsychotic Agents/administration & dosage , Aripiprazole/administration & dosage , Aripiprazole/therapeutic use , Drug Therapy, Combination , Humans , Male , Middle Aged , Monoamine Oxidase Inhibitors/administration & dosage , Phenelzine/administration & dosage , Phenelzine/therapeutic use , Quetiapine Fumarate/administration & dosage , Quetiapine Fumarate/therapeutic use
13.
Exp Cell Res ; 340(2): 227-37, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26748182

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) are capable of unlimited self-renewal and can generate nearly all cells in the body. Changes induced by different LSD1 activities on the regulation of hiPSC self-renewal and differentiation and the mechanism underlying such changes were determined. We used two different LSD1 inhibitors (phenelzine sulfate and tranylcypromine) and RNAi technique to inhibit LSD1 activity, and we obtained hiPSCs showing 71.3%, 53.28%, and 31.33% of the LSD1 activity in normal hiPSCs. The cells still maintained satisfactory self-renewal capacity when LSD1 activity was at 71.3%. The growth rate of hiPSCs decreased and cells differentiated when LSD1 activity was at approximately 53.28%. The hiPSCs were mainly arrested in the G0/G1 phase and simultaneously differentiated into endodermal tissue when LSD1 activity was at 31.33%. Teratoma experiments showed that the downregulation of LSD1 resulted in low teratoma volume. When LSD1 activity was below 50%, pluripotency of hiPSCs was impaired, and the teratomas mainly comprised endodermal and mesodermal tissues. This phenomenon was achieved by regulating the critical balance between histone methylation and demethylation at regulatory regions of several key pluripotent and developmental genes.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Cell Self Renewal , Histone Demethylases/metabolism , Induced Pluripotent Stem Cells/cytology , Cell Line , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/genetics , Histones/metabolism , Humans , Methylation , Phenelzine/pharmacology , RNA Interference , Tranylcypromine/pharmacology
14.
Zoolog Sci ; 34(2): 112-121, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28397602

ABSTRACT

To study the effects of post-glacial isolation by islands on population genetic diversity and differentiation of the large Japanese field mouse, Apodemus speciosus, we examined partial nucleotide sequences of the mitochondrial Dloop region (ca. 300 bp) in 231 individuals collected from islands in the Seto Inland Sea and adjacent regions on Honshu and Shikoku Islands in the western part of the Japanese archipelago. Molecular phylogenetic and network analyses showed that haplotypes in each island tended to form monophyletic groups, while those in Honshu and Shikoku (the major Japanese islands) showed scattered relationships and were connected with island haplotypes. These observations suggest that a set of Honshu and Shikoku haplotypes became the ancestral lineages of the island population. No gene flow was detected among island populations, indicating that independent evolution occurred on each island, without the influence of human activities, since the establishment of the islands in the Holocene. Population genetic diversities on each island were lower than those on Honshu and Shikoku. Comparison between genetic diversity and island area size showed positive correlations and supported the suggestion that genetic drift is a major factor that shaped the current haplotype constitution of the islands in the Seto Inland Sea.


Subject(s)
Animal Distribution/physiology , DNA, Mitochondrial/genetics , Genetic Variation , Islands , Murinae/genetics , Animals , Base Sequence , Haplotypes , Japan , Phenelzine , Phylogeny , Protein Binding
15.
J Neurochem ; 138(2): 328-38, 2016 07.
Article in English | MEDLINE | ID: mdl-27060873

ABSTRACT

Currently there are no effective therapies available for the excruciating neuropathic pain that develops after spinal cord injuries (SCI). As such, a great deal of effort is being put into the investigation of novel therapeutic targets that can alleviate this pain. One such target is acrolein, a highly reactive aldehyde produced as a byproduct of oxidative stress and inflammation that is capable of activating the transient receptor potential ankyrin 1 (TRPA1) cation channel, known to be involved in the transmission and propagation of chronic neuropathic pain. One anti-acrolein agent, hydralazine, has already been shown to reduce neuropathic pain behaviors and offer neuroprotection after SCI. This study investigates another acrolein scavenger, phenelzine, for its possible role of alleviating sensory hypersensitivity through acrolein suppression. The results show that phenelzine is indeed capable of attenuating neuropathic pain behaviors in acute, delayed, and chronic administration schedules after injury in a rat model of SCI. In addition, upon the comparison of hydralazine to phenelzine, both acrolein scavengers displayed a dose-dependent response in the reduction of acrolein in vivo. Finally, phenelzine proved capable of providing locomotor function recovery and neuroprotection of spinal cord tissue when administered immediately after injury for 2 weeks. These results indicate that phenelzine may be an effective treatment for neuropathic pain after SCI and likely a viable alternative to hydralazine. We have shown that phenelzine can attenuate neuropathic pain behavior in acute, delayed, and chronic administration in post-SCI rats. This was accompanied by a dose-dependent reduction in an acrolein metabolite in urine and an acrolein adduct in spinal cord tissue, and the suppression of TRPA1 over-expression in central and peripheral locations post-trauma. Acrolein scavenging might be a novel therapeutic strategy to reduce post-SCI neuropathic pain.


Subject(s)
Hyperalgesia/drug therapy , Neuralgia/drug therapy , Phenelzine/pharmacology , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Animals , Behavior, Animal/drug effects , Contusions/drug therapy , Disease Models, Animal , Hydralazine/toxicity , Male , Neuralgia/metabolism , Rats, Sprague-Dawley , Spinal Cord Injuries/metabolism
16.
J Neuroinflammation ; 13(1): 142, 2016 06 10.
Article in English | MEDLINE | ID: mdl-27282914

ABSTRACT

BACKGROUND: Chronic neuropathic pain is a common symptom of multiple sclerosis (MS). MOG35-55-induced experimental autoimmune encephalomyelitis (EAE) has been used as an animal model to investigate the mechanisms of pain in MS. Previous studies have implicated sensitization of spinal nociceptive networks in the pathogenesis of pain in EAE. However, the involvement of supraspinal sites of nociceptive integration, such as the primary somatosensory cortex (S1), has not been defined. We therefore examined functional, structural, and immunological alterations in S1 during the early stages of EAE, when pain behaviors first appear. We also assessed the effects of the antidepressant phenelzine (PLZ) on S1 alterations and nociceptive (mechanical) sensitivity in early EAE. PLZ has been shown to restore central nervous system (CNS) tissue concentrations of GABA and the monoamines (5-HT, NA) in EAE. We hypothesized that PLZ treatment would also normalize nociceptive sensitivity in EAE by restoring the balance of excitation and inhibition (E-I) in the CNS. METHODS: We used in vivo flavoprotein autofluorescence imaging (FAI) to assess neural ensemble responses in S1 to vibrotactile stimulation of the limbs in early EAE. We also used immunohistochemistry (IHC), and Golgi-Cox staining, to examine synaptic changes and neuroinflammation in S1. Mechanical sensitivity was assessed at the clinical onset of EAE with Von Frey hairs. RESULTS: Mice with early EAE exhibited significantly intensified and expanded FAI responses in S1 compared to controls. IHC revealed increased vesicular glutamate transporter (VGLUT1) expression and disrupted parvalbumin+ (PV+) interneuron connectivity in S1 of EAE mice. Furthermore, peri-neuronal nets (PNNs) were significantly reduced in S1. Morphological analysis of excitatory neurons in S1 revealed increased dendritic spine densities. Iba-1+ cortical microglia were significantly elevated early in the disease. Chronic PLZ treatment was found to normalize mechanical thresholds in EAE. PLZ also normalized S1 FAI responses, neuronal morphologies, and cortical microglia numbers and attenuated VGLUT1 reactivity-but did not significantly attenuate the loss of PNNs. CONCLUSIONS: These findings implicate a pro-excitatory shift in the E-I balance of the somatosensory CNS, arising early in the pathogenesis EAE and leading to large-scale functional and structural plasticity in S1. They also suggest a novel antinociceptive effect of PLZ treatment.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/complications , Neuralgia/etiology , Neuralgia/pathology , Pain Threshold/physiology , Somatosensory Cortex/pathology , Synapses/metabolism , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Freund's Adjuvant/toxicity , Hyperalgesia/etiology , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein/toxicity , Neuralgia/drug therapy , Neurons/cytology , Neurons/metabolism , Neurons/ultrastructure , Pain Measurement/drug effects , Pain Threshold/drug effects , Parvalbumins/metabolism , Peptide Fragments/immunology , Peptide Fragments/toxicity , Phenelzine/pharmacology , Phenelzine/therapeutic use , Plant Lectins/metabolism , Receptors, N-Acetylglucosamine/metabolism , Somatosensory Cortex/drug effects , Somatosensory Cortex/ultrastructure , Synapses/pathology , Synapses/ultrastructure
17.
Aust N Z J Psychiatry ; 55(8): 831-832, 2021 08.
Article in English | MEDLINE | ID: mdl-33040569

Subject(s)
Phenelzine , Humans
18.
Australas Psychiatry ; 24(4): 371-3, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26917855

ABSTRACT

OBJECTIVE: The place of monoamine oxidase inhibitors (MAOIs) in psychiatry is reviewed, and the question posed as to whether they are now justifiably disregarded by prescribers. METHOD: Multiple databases (PubMed, Medline, Embase, Cochrane) were interrogated to provide an overview regarding the use, efficacy and toxicity of MAOIs. Data regarding funded use of these agents in New Zealand were obtained from PHARMAC. RESULTS: Evidence supports the use of MAOIs in major depressive disorder, certain anxiety disorders and, to lesser extent, bipolar depression. Older non-selective agents, such as phenelzine and tranylcypromine, have distinctive efficacy in 'atypical' and treatment-resistant depression, but at the cost of serious tolerability problems. Their relegation and perception by clinicians as 'last resort' medications - if considered at all - has occurred in the context of various concerns, notably dietary restrictions, potential adverse drug interactions and the usual requirement for divided doses. CONCLUSIONS: Sufficient evidence supports consideration of MAOIs in treatment-refractory and atypical depressive disorders, and in social anxiety disorder. Psychiatrists in training need to gain experience in using these agents.


Subject(s)
Depressive Disorder, Treatment-Resistant/drug therapy , Monoamine Oxidase Inhibitors/therapeutic use , Phenelzine/therapeutic use , Phobia, Social/drug therapy , Tranylcypromine/therapeutic use , Anxiety Disorders/drug therapy , Bipolar Disorder/drug therapy , Depressive Disorder, Major/drug therapy , Humans , Monoamine Oxidase Inhibitors/adverse effects , Phenelzine/adverse effects , Randomized Controlled Trials as Topic , Tranylcypromine/adverse effects
19.
Int J Neuropsychopharmacol ; 19(1)2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26316187

ABSTRACT

BACKGROUND: Monoamine oxidase inhibitors (MAOIs) are being developed for major depressive disorder, Alzheimer's, and Parkinson's Disease. Newer MAOIs have minimal sensitivity to tyramine, but a key limitation for optimizing their development is that standards for in vivo monoamine oxidase-A (MAO-A) occupancy in humans are not well established. The objectives were to determine the dose-occupancy relationship of moclobemide and the occupancy of phenelzine at typical clinical dosing. METHODS: Major depressive episode (MDE) subjects underwent [(11)C]harmine positron emission tomography scanning prior to and following 6 weeks of treatment with moclobemide or phenelzine. RESULTS: Mean brain MAO-A occupancies were 74.23±8.32% for moclobemide at 300-600 mg daily (n = 11), 83.75±5.52% for moclobemide at 900-1200 mg daily (n = 9), and 86.82±6.89% for phenelzine at 45-60 mg daily (n = 4). The regional dose-occupancy relationship of moclobemide fit a hyperbolic function [F(x) = a(x/[b + x]); F(1,18) = 5.57 to 13.32, p = 0.002 to 0.03, mean 'a': 88.62±2.38%, mean 'b': 69.88±4.36 mg]. Multivariate analyses of variance showed significantly greater occupancy of phenelzine (45-60mg) and higher-dose moclobemide (900-1200 mg) compared to lower-dose moclobemide [300-600 mg; F(7,16) = 3.94, p = 0.01]. CONCLUSIONS: These findings suggest that for first-line MDE treatment, daily moclobemide doses of 300-600mg correspond to a MAO-A occupancy of 74%, whereas for treatment-resistant MDE, either phenelzine or higher doses of moclobemide correspond to a MAO-A occupancy of at least 84%. Therefore, novel MAO inhibitor development should aim for similar thresholds. The findings provide a rationale in treatment algorithm design to raise moclobemide doses to inhibit more MAO-A sites, but suggest switching from high-dose moclobemide to phenelzine is best justified by binding to additional targets.


Subject(s)
Brain/drug effects , Moclobemide/pharmacology , Monoamine Oxidase Inhibitors/pharmacokinetics , Monoamine Oxidase/metabolism , Phenelzine/pharmacology , Adult , Brain/diagnostic imaging , Carbon Isotopes/pharmacokinetics , Dose-Response Relationship, Drug , Female , Harmine/pharmacokinetics , Healthy Volunteers , Humans , Male , Middle Aged , Multivariate Analysis , Positron-Emission Tomography , Protein Binding/drug effects , Young Adult
20.
J Biol Chem ; 288(44): 31517-27, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24043630

ABSTRACT

Proteins misfolded in the endoplasmic reticulum (ER) are cleared by the ubiquitin-dependent proteasome system in the cytosol, a series of events collectively termed ER-associated degradation (ERAD). It was previously shown that SEL1L, a partner protein of the E3 ubiquitin ligase HRD1, is required for degradation of misfolded luminal proteins (ERAD-Ls substrates) but not misfolded transmembrane proteins (ERAD-Lm substrates) in both mammalian and chicken DT40 cells. Here, we analyzed ATF6, a type II transmembrane glycoprotein that serves as a sensor/transducer of the unfolded protein response, as a potential ERAD-Lm substrate in DT40 cells. Unexpectedly, degradation of endogenous ATF6 and exogenously expressed chicken and human ATF6 by the proteasome required SEL1L. Deletion analysis revealed that the luminal region of ATF6 is a determinant for SEL1L-dependent degradation. Chimeric analysis showed that the luminal region of ATF6 confers SEL1L dependence on type I transmembrane protein as well. In contrast, degradation of other known type I ERAD-Lm substrates (BACE457, T-cell receptor-α, CD3-δ, and CD147) did not require SEL1L. Thus, ATF6 represents a novel type of ERAD-Lm substrate requiring SEL1L for degradation despite its transmembrane nature. In addition, endogenous ATF6 was markedly stabilized in wild-type cells treated with kifunensine, an inhibitor of α1,2-mannosidase in the ER, indicating that degradation of ATF6 requires proper mannose trimming. Our further analyses revealed that the five ERAD-Lm substrates examined are classified into three subgroups based on their dependence on mannose trimming and SEL1L. Thus, ERAD-Lm substrates are degraded through much more diversified mechanisms in higher eukaryotes than previously thought.


Subject(s)
Activating Transcription Factor 6/metabolism , Endoplasmic Reticulum-Associated Degradation/physiology , Mannose/metabolism , Mannosidases/metabolism , Proteins/metabolism , Activating Transcription Factor 6/genetics , Alkaloids/pharmacology , Animals , Antidepressive Agents/pharmacology , Cell Line , Chickens , Endoplasmic Reticulum-Associated Degradation/drug effects , Enzyme Inhibitors/pharmacology , Mannose/genetics , Mannosidases/antagonists & inhibitors , Mannosidases/genetics , Phenelzine/pharmacology , Protein Stability/drug effects , Proteins/genetics , Proteolysis/drug effects , Substrate Specificity/drug effects , Substrate Specificity/physiology
SELECTION OF CITATIONS
SEARCH DETAIL