1
|
Zhang Y, Zhang W, Yu L, Shi Y, Xu M, Wang H, Li C, Tian J. The TAAR1 Agonist PCC0105004 Regulates Amygdala Synaptic Plasticity to Alleviate Anxiety-Like Behaviors in Rats. Pharmacol Res Perspect 2025; 13:e70068. [PMID: 40186385 PMCID: PMC11971484 DOI: 10.1002/prp2.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/26/2024] [Accepted: 01/10/2025] [Indexed: 04/07/2025] Open
Abstract
Anxiety disorder is a persistent, widespread, and intractable mood disorder, and the available pharmacotherapies have limited efficacy with significant side effects. Trace amine-associated receptor 1 (TAAR1) is an emerging drug target for neuropsychiatric disorders. This study examined the effects and underlying mechanisms of a novel TAAR1 agonist, PCC0105004, in a rat model of CUMS-induced anxiety-like behavior. The elevated zero maze and open field tests test were employed to evaluate the anti-anxiety-like activity of PCC0105004. PCC0105004 dose-dependently attenuated anxiety-like behaviors in rats without affecting spontaneous activity. Morphologically, PCC0104005 decreased the density of dendritic spines in the amygdala. For the mechanistic studies, whole-genome transcriptomic analysis revealed significant differences in the patterns of amygdala gene expression in the CUMS-induced anxiety rat model. These transcriptomic data were further confirmed by using RT-qPCR and western blotting, further revealing alterations associated with genes (Col1a1, DCN, Ewsr1) known to regulate synaptic plasticity, and PCC0105004 was able to reverse these changes. These results suggest that PCC0105004 is a promising anxiolytic candidate for pharmacotherapy of anxiety and warrants further examination and development.
Collapse
Affiliation(s)
- Yingtian Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai UniversityYantaiPeople's Republic of China
| | - Wei Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai UniversityYantaiPeople's Republic of China
| | - Linyao Yu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai UniversityYantaiPeople's Republic of China
| | - Yaoqin Shi
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai UniversityYantaiPeople's Republic of China
| | - Min Xu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai UniversityYantaiPeople's Republic of China
| | - Hui Wang
- State Key Laboratory of Advanced Drug Delivery and Release SystemsYantaiShandongPeople's Republic of China
| | - Chunmei Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai UniversityYantaiPeople's Republic of China
- State Key Laboratory of Advanced Drug Delivery and Release SystemsYantaiShandongPeople's Republic of China
| | - Jingwei Tian
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of EducationCollaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai UniversityYantaiPeople's Republic of China
- State Key Laboratory of Advanced Drug Delivery and Release SystemsYantaiShandongPeople's Republic of China
| |
Collapse
|
2
|
Van Dongen HPA, Leary EB, Drake C, Bogan R, Jaeger J, Rosenberg R, Streicher C, Tabuteau H. Results of the Solriamfetol's Effect on Cognitive Health in Apnea Participants During a Randomized Placebo-Controlled Study (SHARP): A Randomized Placebo-Controlled Double-Blind Repeated-Measures Crossover Phase IV Clinical Trial of the Effect of the Wake-Promoting Agent Solriamfetol on Cognitive Function in OSA With Excessive Daytime Sleepiness and Cognitive Impairment. Chest 2025; 167:863-875. [PMID: 39528111 DOI: 10.1016/j.chest.2024.10.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/13/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND OSA causes episodes of fragmented sleep and intermittent hypoxia and leads to excessive daytime sleepiness (EDS). Deficits in cognitive function are a troublesome symptom in patients with OSA and EDS. RESEARCH QUESTION How does solriamfetol affect cognitive function in patients with cognitive impairment associated with OSA and EDS? STUDY DESIGN AND METHODS Solriamfetol's Effect on Cognitive Health in Apnea Participants During a Randomized Placebo-Controlled Study (SHARP) was a phase IV, randomized double-blind placebo-controlled crossover trial. Participants (N = 59) were randomized to receive placebo or solriamfetol (75 mg/d for 3 days, then 150 mg/d) for 2 weeks, with crossover separated by a 1-week washout period. Efficacy measures included the Coding subtest, comparable to the Digit Symbol Substitution Test (DSST), of the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS), the British Columbia Cognitive Complaints Inventory (BC-CCI), the Patient Global Impression of Severity (PGI-S), and the Epworth Sleepiness Scale (ESS). The primary end point was change from baseline in average postdose DSST RBANS scores. Secondary end points were changes from baseline in BC-CCI, PGI-S, ESS, and DSST RBANS scores at 2, 4, 6, and 8 hours' postdose. Safety was monitored by assessment of treatment-emergent adverse events. RESULTS Solriamfetol was shown to significantly improve postdose average DSST RBANS scores compared with placebo (P = .009; effect size [Cohen's d], 0.37). When evaluated at each 2-hour time point, cognitive function was significantly improved at 2, 6, and 8 hours after dosing (all, P < .05). During solriamfetol treatment, there were significant improvements in BC-CCI (P = .002; d = 0.45), PGI-S (P = .034; d = 0.29), and ESS (P = .004; d = 0.40) compared with placebo. The most common treatment-emergent adverse events were nausea (7%) and anxiety (3%). INTERPRETATION SHARP showed that solriamfetol can improve objective and subjective measures of cognitive function in patients with cognitive impairment associated with OSA and EDS. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov; No.: NCT04789174; URL: www. CLINICALTRIALS gov and EudraCT; No.: 2020-004243-92; URL: https://eudract.ema.europa.eu.
Collapse
Affiliation(s)
- Hans P A Van Dongen
- Department of Translational Medicine and Physiology & Sleep and Performance Research Center, Washington State University, Spokane, WA
| | | | - Christopher Drake
- Henry Ford Hospital Sleep Disorders and Research Center, Detroit, MI
| | - Richard Bogan
- SleepMed, Inc, Columbia, SC; University of South Carolina School of Medicine, Columbia, SC
| | - Judith Jaeger
- CognitionMetrics, LLC, Stamford, CT; Albert Einstein College of Medicine, Bronx, NY
| | | | | | | |
Collapse
|
3
|
Winter Y, Mayer G, Benes H, Burghaus L, Floam S, Parks GS, Kallweit U. Patients with obstructive sleep apnea in Germany. Sleep Breath 2025; 29:112. [PMID: 40014191 PMCID: PMC11868153 DOI: 10.1007/s11325-025-03275-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/28/2025]
Abstract
PURPOSE Solriamfetol is approved for use in the European Union to treat excessive daytime sleepiness (EDS) associated with obstructive sleep apnea (OSA). SURWEY characterized real-world evidence regarding physician initiation and titration strategies and patient experiences with solriamfetol. We report SURWEY data for patients with OSA and EDS in Germany (N = 83). METHODS SURWEY was a retrospective chart review conducted among physicians in Germany. Eligible patients were age ≥ 18 years who reached a stable solriamfetol dose and completed ≥ 6 weeks of treatment. Patients were grouped by solriamfetol initiation strategy: changeover, add-on, new-to-therapy. RESULTS Patients' mean (SD) age was 49 (14) years. New-to-therapy was the most common initiation strategy. Solriamfetol was initiated at 37.5 mg/day in most patients (n = 57, 69%) and titrated in 53 patients (64%); 30 (57%) completed titration within 2 weeks. In a post-hoc analysis, mean (SD) Epworth Sleepiness Scale (ESS) score was 16.0 (3.2) at baseline and decreased by 5.4 (3.6) at final follow-up (~ 16 weeks; p <.001). Improvement in patient- and physician-rated EDS was reported by ~ 90% of patients. Most patients (55%) reported effects of solriamfetol lasting ≥ 8 h; 91% of patients reported no change in nighttime sleep quality. The most frequent adverse events were headache (8%), decreased appetite (7%), and insomnia (6%). CONCLUSION Most patients in this study were new to therapy. Solriamfetol was typically initiated at 37.5 mg/day; titration was common. ESS scores improved with solriamfetol treatment, and most patients self-reported improvement in EDS symptoms. Common adverse events were consistent with those reported in previous clinical trials.
Collapse
Affiliation(s)
- Yaroslav Winter
- Department of Neurology, University of Saarland, Homburg, Germany.
- Department of Neurology, Philipps University Marburg, Marburg, Germany.
- Department of Neurology, University of Saarland, Homburg, Germany, Kirrberger Str, 66421.
| | - Geert Mayer
- Department of Neurology, Philipps University Marburg, Marburg, Germany
- Hephata Klinik, Schwalmstadt, Germany
| | - Heike Benes
- Somni bene GmbH Institut für Medizinische Forschung und Schlafmedizin Schwerin GmbH, Schwerin, Germany
| | - Lothar Burghaus
- Department of Neurology, Heilig Geist-Hospital, Cologne, Germany
| | | | | | - Ulf Kallweit
- Center for Biomedical Education and Research, and Professorship for Narcolepsy and Hypersomnolence Research, Department of Medicine, University Witten/Herdecke, Witten, Germany
| |
Collapse
|
4
|
Apryatin SA. The Neurometabolic Function of the Dopamine-Aminotransferase System. Metabolites 2025; 15:21. [PMID: 39852364 PMCID: PMC11767981 DOI: 10.3390/metabo15010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES The neurometabolic function is controlled by a complex multi-level physiological system that includes neurochemical, hormonal, immunological, sensory, and metabolic components. Functional disorders of monoamine systems are often detected in clinical practice together with metabolic dysfunctions. An important part of the mentioned pathological conditions are associated with disturbances in protein metabolism, some of the most important biomarkers which are aminotransferases and transcription factors that regulate and direct the most important metabolic reactions. Another important part of energy metabolism is the dopamine-mediated regulation of protein metabolism. METHODS The review describes research results into the dopamine-mediated mechanism of metabolic regulation in humans and animals. Particular attention is paid to the neurometabolic mechanisms of protein metabolism. RESULTS The dopamine-aminotransferase system of the energy metabolism regulation is a separate, independent, regulatory and diagnostically significant biochemical pathway controlled by the hormonal system, the key hormone is cortisol, the key neurotransmitter is dopamine, the key transcription factor is CREB, and the key regulatory enzymes are alanine aminotransferase, aspartate aminotransferase, and tyrosine aminotransferase. CONCLUSIONS This review presents an original study describing the discovery of a new regulatory mechanism for neurometabolic physiological function in humans and animals. A key part of this mechanism is the dopamine-aminotransferase system.
Collapse
Affiliation(s)
- Sergey A Apryatin
- Institute of Translational Biomedicine, Saint Petersburg State University, 199034 Saint Petersburg, Russia
| |
Collapse
|
5
|
Biso L, Carli M, Scarselli M, Longoni B. Overview of Novel Antipsychotic Drugs: State of the Art, New Mechanisms, and Clinical Aspects of Promising Compounds. Biomedicines 2025; 13:85. [PMID: 39857669 PMCID: PMC11763187 DOI: 10.3390/biomedicines13010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Antipsychotic medications are a vast class of drugs used for the treatment of psychotic disorders such as schizophrenia. Although numerous compounds have been developed since their introduction in the 1950s, several patients do not adequately respond to current treatments, or they develop adverse reactions that cause treatment discontinuation. Moreover, in the past few decades, discoveries in the pathophysiology of psychotic disorders have opened the way for experimenting with novel compounds that have alternative mechanisms of action, with some of them showing promising results in early trials. The scope of this review was to summarize the novel antipsychotics developed, their current experimental status, and their mechanisms of action. In particular, we analyzed the main classes of investigational antipsychotics, such as monoamine, glutamate, acetylcholine, cannabinoid receptor modulators, enzyme inhibitors, ion channel modulators, and mixed receptor modulators. In addition, the safety profiles and adverse effects of these drugs were carefully evaluated, considering the relevance of these aspects for patients' drug adherence and quality of life, especially in the long-term treatment. Lastly, we tried to understand which compounds have greater potential to be approved by the principal drug regulatory agencies in the next years and if they could be used for diseases other than psychotic disorders.
Collapse
Affiliation(s)
| | | | | | - Biancamaria Longoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (L.B.); (M.C.); (M.S.)
| |
Collapse
|
6
|
Zhukov IS, Alnefeesi Y, Krotova NA, Nemets VV, Demin KA, Karpenko MN, Budygin EA, Kanov EV, Kalueff AV, Shabanov PD, Bader M, Alenina N, Gainetdinov RR. Trace amine-associated receptor 1 agonist reduces aggression in brain serotonin-deficient tryptophan hydroxylase 2 knockout rats. Front Psychiatry 2024; 15:1484925. [PMID: 39748904 PMCID: PMC11693706 DOI: 10.3389/fpsyt.2024.1484925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction Aggression and self-harm disproportionately occur in youths preoccupied with social status tracking. These pathological conditions are linked to a serotonin (5-HT) deficit in the brain. Ablation of 5-HT biosynthesis by tryptophan hydroxylase 2 knockout (TPH2-KO) increases aggression in rodents. Remarkably, deletion of the trace amine-associated receptor 1 (TAAR1) results in the same consequences. Unlike the nuanced dynamics of social status cues in young people, the social ranks of rats mainly advance when they dominate larger opponents in combat. Methods This study explored whether the potent TAAR1 agonist RO5263397 reduces aggression caused by 5-HT depletion, and whether social rank advancement motivates this aggression. The resident-intruder paradigm was applied with larger and smaller intruders to evaluate whether social rank advancement motivates aggressive behaviors in TPH2-KO rats. Results When a smaller intruder was introduced, 5-HT-deficient rats did not differ from wild type littermates. However, when the intruders were larger, the mutants extended their aggressive efforts, refusing to submit. Importantly, RO5263397 selectively abolished this abnormal form of aggression in TPH2-KO rats. Discussion Results supported social rank advancement as the main incentive. These data also suggest that TAAR1 is a promising target for the development of new treatments for aggression; independent data also support this conclusion.
Collapse
Affiliation(s)
- Ilya S. Zhukov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Institute of Experimental Medicine, Saint Petersburg, Russia
| | - Yazen Alnefeesi
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | | | - Vsevolod V. Nemets
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Konstantin A. Demin
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | | | - Evgeny A. Budygin
- Department of Neurobiology, Sirius University of Science and Technology, Sirius, Russia
| | - Evgeny V. Kanov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- St. Petersburg University Hospital, St. Petersburg State University, St. Petersburg, Russia
| | - Allan V. Kalueff
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- Department of Neurobiology, Sirius University of Science and Technology, Sirius, Russia
- Department of Biosciences and Bioinformatics, School of Science, Xi’an Jiaotong-Liverpool University, Suzhou, China
- Suzhou Municipal Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi’an Jiaotong-Liverpool University, Suzhou, China
| | | | - Michael Bader
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Natalia Alenina
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
- St. Petersburg University Hospital, St. Petersburg State University, St. Petersburg, Russia
| |
Collapse
|
7
|
Park S, Heu J, Hoener MC, Kilduff TS. Wakefulness Induced by TAAR1 Partial Agonism in Mice Is Mediated Through Dopaminergic Neurotransmission. Int J Mol Sci 2024; 25:11351. [PMID: 39518904 PMCID: PMC11547084 DOI: 10.3390/ijms252111351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Trace amine-associated receptor 1 (TAAR1) is a negative regulator of dopamine (DA) release. The partial TAAR1 agonist RO5263397 promotes wakefulness and suppresses NREM and REM sleep in rodents and non-human primates. We tested the hypothesis that the TAAR1-mediated effects on sleep/wake regulation were due, in part, to DA release. Male C57BL6/J mice (n = 8) were intraperitoneally administered the D1R antagonist SCH23390, the D2R antagonist eticlopride, a combination of D1R + D2R antagonists, or saline at ZT5.5, followed 30 min later by RO5263397 or vehicle per os. EEG, EMG, subcutaneous temperature, and activity were recorded across the 8 treatments and sleep architecture was analyzed for 6 h post-dosing. As described previously, RO5263397 increased wakefulness and delayed NREM and REM sleep onset. D1, D2, and D1 + D2 pretreatment reduced RO5263397-induced wakefulness for 1-2 h after dosing but only the D1 antagonist significantly reduced the TAAR1-mediated increase in NREM latency. Neither the D1 nor the D2 antagonist affected the TAAR1-mediated suppression of REM sleep. These results suggest that, whereas the TAAR1 effects on wakefulness are mediated, in part, through the D2R, D1R activation plays a role in reversing the TAAR1-mediated increase in NREM sleep latency. In contrast, the TAAR1-mediated suppression of REM sleep appears not to involve D1R or D2R mechanisms.
Collapse
Affiliation(s)
- Sunmee Park
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA 94025, USA; (S.P.); (J.H.)
| | - Jasmine Heu
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA 94025, USA; (S.P.); (J.H.)
| | - Marius C. Hoener
- Neuroscience and Rare Diseases Discovery & Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland;
| | - Thomas S. Kilduff
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA 94025, USA; (S.P.); (J.H.)
| |
Collapse
|
8
|
Park S, Heu J, Hoener MC, Kilduff TS. Wakefulness Induced by TAAR1 Partial Agonism is Mediated Through Dopaminergic Neurotransmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612122. [PMID: 39314371 PMCID: PMC11419104 DOI: 10.1101/2024.09.09.612122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Trace amine-associated receptor 1 (TAAR1) is known to negatively regulate dopamine (DA) release. The partial TAAR1 agonist RO5263397 promotes wakefulness and suppresses NREM and REM sleep in mice, rats, and non-human primates. We tested the hypothesis that the TAAR1-mediated effects on sleep/wake were due, at least in part, to DA release. Male C57BL6/J mice (n=8) were intraperitoneally administered the D1R antagonist SCH23390, the D2R antagonist eticlopride, a combination of D1R+D2R antagonists or saline at ZT5.5, followed 30 min later by RO5263397 or vehicle (10% DMSO in DI water) at ZT6 per os. EEG, EMG, subcutaneous temperature, and activity were recorded in each mouse across the 8 treatment conditions and sleep architecture was analyzed for 6 hours post-dosing. Consistent with our previous reports, RO5263397 increased wakefulness as well as the latency to NREM and REM sleep. D1, D2, and D1+D2 pretreatment reduced RO5263397-induced wakefulness during the first 1-2 hours after dosing, but only the D1+D2 combination attenuated the wake-promoting effect of RO5263397 from ZT6-8, mostly by increasing NREM sleep. Although D1+D2 antagonism blocked the wake-promoting effect of RO5263397, only the D1 antagonist significantly reduced the TAAR1-mediated increase in NREM latency. Neither the D1 nor the D2 antagonist affected TAAR1-mediated suppression of REM sleep. These results suggest that, whereas TAAR1 effects on wakefulness are mediated in part through the D2R, D1R activation plays a role in reversing the TAAR1-mediated increase in NREM sleep latency. By contrast, TAAR1-mediated suppression of REM sleep appears not to involve D1R or D2R mechanisms.
Collapse
Affiliation(s)
- Sunmee Park
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA
| | - Jasmine Heu
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA
| | - Marius C. Hoener
- Neuroscience, Ophthalmology and Rare Diseases DTA, pRED, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Thomas S. Kilduff
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, CA
| |
Collapse
|
9
|
Alnefeesi Y, Sukhanov I, Gainetdinov RR. Ligands of the trace amine-associated receptors (TAARs): A new class of anxiolytics. Pharmacol Biochem Behav 2024; 242:173817. [PMID: 39002806 DOI: 10.1016/j.pbb.2024.173817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/15/2024]
Abstract
Most cases of anxiety are currently treated with either benzodiazepines or serotonin reuptake inhibitors. These drugs carry with them risks for a multitude of side effects, and patient compliance suffers for this reason. There is thus a need for novel anxiolytics, and among the most compelling prospects in this vein is the study of the TAARs. The anxiolytic potential of ulotaront, a full agonist at the human TAAR1, is currently being investigated in patients with generalized anxiety disorder. Irrespective of whether this compound succeeds in clinical trials, a growing body of preclinical literature underscores the relevance of modulating the TAARs in anxiety. Multiple behavioral paradigms show anxiolytic-like effects in rodents, possibly due to increased neurogenesis and plasticity, in addition to a panoply of interactions between the TAARs and other systems. Crucially, multiple lines of evidence suggest that the TAARs, particularly TAAR1, TAAR2, and TAAR5, are expressed in the extended amygdala and hippocampus. These regions are central in the actuation of anxiety, and are particularly susceptible to neurogenic and neuroplastic effects which the TAARs are now known to regulate. The TAARs also regulate the dopamine and serotonin systems, both of which are implicated in anxiety. Ligands of the TAARs may thus constitute a new class of anxiolytics.
Collapse
Affiliation(s)
- Yazen Alnefeesi
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Ilya Sukhanov
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, 197022 St. Petersburg, Russia
| | - Raul R Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; St. Petersburg University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia.
| |
Collapse
|
10
|
Yang Y, Qiao X, Yu S, Zhao X, Jin Y, Liu R, Li J, Wang L, Song L. A trace amine associated receptor mediates antimicrobial immune response in the oyster Crassostrea gigas. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 156:105171. [PMID: 38537729 DOI: 10.1016/j.dci.2024.105171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/21/2024] [Accepted: 03/24/2024] [Indexed: 04/01/2024]
Abstract
Trace amine-associated receptors (TAARs) are a class of G protein-coupled receptors, playing an immunomodulatory function in the neuroinflammatory responses. In the present study, a TAAR homologue with a 7tm_classA_rhodopsin-like domain (designated as CgTAAR1L) was identified in oyster Crassostrea gigas. The abundant CgTAAR1L transcripts were detected in visceral ganglia and haemocytes compared to other tissues, which were 55.35-fold and 32.95-fold (p < 0.01) of those in adductor muscle, respectively. The mRNA expression level of CgTAAR1L in haemocytes significantly increased and reached the peak level at 3 h after LPS or Poly (I:C) stimulation, which was 4.55-fold and 12.35-fold of that in control group, respectively (p < 0.01). After the expression of CgTAAR1L was inhibited by the injection of its targeted siRNA, the mRNA expression levels of interleukin17s (CgIL17-1, CgIL17-5 and CgIL17-6), and defensin (Cgdefh1) significantly decreased at 3 h after LPS stimulation, which was 0.51-fold (p < 0.001), 0.39-fold (p < 0.01), 0.48-fold (p < 0.05) and 0.41-fold (p < 0.05) of that in the control group, respectively. The nuclear translocation of Cgp65 protein was suppressed in the CgTAAR1L-RNAi oysters. Furthermore, the number of Vibrio splendidus in the haemolymph of CgTAAR1L-RNAi oysters significantly increased (4.11-fold, p < 0.001) compared with that in the control group. In contrast, there was no significant difference in phagocytic rate of haemocytes to V. splendidus in the CgTAAR1L-RNAi oysters. These results indicated that CgTAAR1L played an important role in the immune defense against bacterial infection by inducing the expressions of interleukin and defensin.
Collapse
Affiliation(s)
- Yuehong Yang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Xue Qiao
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Simiao Yu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Xinyu Zhao
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Yuhao Jin
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Rui Liu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Jie Li
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| |
Collapse
|
11
|
Zhang Y, Zhang XQ, Niu WP, Sun M, Zhang Y, Li JT, Si TM, Su YA. TAAR1 in dentate gyrus is involved in chronic stress-induced impairments in hippocampal plasticity and cognitive function. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110995. [PMID: 38514038 DOI: 10.1016/j.pnpbp.2024.110995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/09/2024] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
Multiple lines of evidence suggest that the trace amine-associated receptor 1 (TAAR1) holds promise as a potential target for stress-related disorders, such as treating major depressive disorder (MDD). The role of TAAR1 in the regulation of adult neurogenesis is recently supported by transcriptomic data. However, it remains unknown whether TAAR1 in dentate gyrus (DG) mediate chronic stress-induced negative effects on hippocampal plasticity and related behavior in mice. The present study consisted of a series of experiments using RNAscope, genetic approaches, behavioral tests, immunohistochemical staining, Golgi-Cox technique to unravel the effects of TAAR1 on alterations of dentate neuronal plasticity and cognitive function in the chronic social defeat stress model. The mice subjected to chronic defeat stress exhibited a noteworthy decrease in the mRNA level of TAAR1 in DG. Additionally, they exhibited compromised social memory and spatial object recognition memory, as well as impaired proliferation and maturation of adult-born dentate granule cells. Moreover, the selective knockout TAAR1 in DG mostly mimicked the cognitive function deficits and neurogenesis impairment induced by chronic stress. Importantly, the administration of the selective TAAR1 partial agonist RO5263397 during stress exposure attenuated the adverse effects of chronic stress on cognitive function, adult neurogenesis, dendritic arborization, and the synapse number of dentate neurons in DG. In summary, our findings suggest that TAAR1 plays a crucial role in mediating the detrimental effects of chronic stress on hippocampal plasticity and cognition. TAAR1 agonists exhibit therapeutic potential for individuals suffering from cognitive impairments associated with MDD.
Collapse
Affiliation(s)
- Yue Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Xian-Qiang Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Wei-Pan Niu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Meng Sun
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, NC 27709, USA
| | - Ji-Tao Li
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Tian-Mei Si
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| | - Yun-Ai Su
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China.
| |
Collapse
|
12
|
Shemiakova TS, Efimova EV, Gainetdinov RR. TAARs as Novel Therapeutic Targets for the Treatment of Depression: A Narrative Review of the Interconnection with Monoamines and Adult Neurogenesis. Biomedicines 2024; 12:1263. [PMID: 38927470 PMCID: PMC11200894 DOI: 10.3390/biomedicines12061263] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/27/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Depression is a common mental illness of great concern. Current therapy for depression is only suitable for 80% of patients and is often associated with unwanted side effects. In this regard, the search for and development of new antidepressant agents remains an urgent task. In this review, we discuss the current available evidence indicating that G protein-coupled trace amine-associated receptors (TAARs) might represent new targets for depression treatment. The most frequently studied receptor TAAR1 has already been investigated in the treatment of schizophrenia, demonstrating antidepressant and anxiolytic properties. In fact, the TAAR1 agonist Ulotaront is currently undergoing phase 2/3 clinical trials testing its safety and efficacy in the treatment of major depressive disorder and generalized anxiety disorder. Other members of the TAAR family (TAAR2, TAAR5, TAAR6, TAAR8, and TAAR9) are not only involved in the innate olfaction of volatile amines, but are also expressed in the limbic brain areas. Furthermore, animal studies have shown that TAAR2 and TAAR5 regulate emotional behaviors and thus may hold promise as potential antidepressant targets. Of particular interest is their connection with the dopamine and serotonin systems of the brain and their involvement in the regulation of adult neurogenesis, known to be affected by the antidepressant drugs currently in use. Further non-clinical and clinical studies are necessary to validate TAAR1 (and potentially other TAARs) as novel therapeutic targets for the treatment of depression.
Collapse
Affiliation(s)
- Taisiia S. Shemiakova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 St. Petersburg, Russia; (T.S.S.); (E.V.E.)
| | - Evgeniya V. Efimova
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 St. Petersburg, Russia; (T.S.S.); (E.V.E.)
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, Saint-Petersburg State University, 199034 St. Petersburg, Russia; (T.S.S.); (E.V.E.)
- Saint-Petersburg University Hospital, Saint-Petersburg State University, 199034 St. Petersburg, Russia
| |
Collapse
|
13
|
Beydoun MA, Beydoun HA, Ashe J, Georgescu MF, Horvath S, Lu A, Zannas AS, Shadyab AH, Jung SY, Wassertheil-Smoller S, Casanova R, Zonderman AB, Brunner RL. Relationships of depression and antidepressant use with epigenetic age acceleration and all-cause mortality among postmenopausal women. Aging (Albany NY) 2024; 16:8446-8471. [PMID: 38809417 PMCID: PMC11164525 DOI: 10.18632/aging.205868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 05/03/2024] [Indexed: 05/30/2024]
Abstract
We investigated relations of depressive symptoms, antidepressant use, and epigenetic age acceleration with all-cause mortality risk among postmenopausal women. Data were analyzed from ≤1,900 participants in the Women's Health Initiative study testing four-way decomposition models. After a median 20.4y follow-up, 1,161 deaths occurred. Approximately 11% had elevated depressive symptoms (EDS+), 7% were taking antidepressant medication at baseline (ANTIDEP+), while 16.5% fell into either category (EDS_ANTIDEP+). Baseline ANTIDEP+, longitudinal transition into ANTIDEP+ and accelerated epigenetic aging directly predicted increased mortality risk. GrimAge DNA methylation age acceleration (AgeAccelGrim) partially mediated total effects of baseline ANTIDEP+ and EDS_ANTIDEP+ on all-cause mortality risk in socio-demographic factors-adjusted models (Pure Indirect Effect >0, P < 0.05; Total Effect >0, P < 0.05). Thus, higher AgeAccelGrim partially explained the relationship between antidepressant use and increased all-cause mortality risk, though only prior to controlling for lifestyle and health-related factors. Antidepressant use and epigenetic age acceleration independently predicted increased all-cause mortality risk. Further studies are needed in varying populations.
Collapse
Affiliation(s)
- May A. Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Hind A. Beydoun
- VA National Center on Homelessness Among Veterans, U.S. Department of Veterans Affairs, Washington, DC 20420, USA
- Department of Management, Policy, and Community Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jason Ashe
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Michael F. Georgescu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Biostatistics, School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ake Lu
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Anthony S. Zannas
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Aladdin H. Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science and Division of Geriatrics, Gerontology, and Palliative Care, Department of Medicine, University of California, San Diego, CA 92093, USA
| | - Su Yon Jung
- Department of Epidemiology, Fielding School of Public Health, Translational Sciences Section, School of Nursing, University of California, Los Angeles, CA 90095, USA
| | - Sylvia Wassertheil-Smoller
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ramon Casanova
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA
| | - Alan B. Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA
| | - Robert L. Brunner
- Department of Family and Community Medicine (Emeritus), School of Medicine, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
14
|
de Bruyn K, Diekman EF, van der Ley CP, van Faassen M, Kema IP. Simultaneous mass spectrometric quantification of trace amines, their precursors and metabolites. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1238:124098. [PMID: 38583227 DOI: 10.1016/j.jchromb.2024.124098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/04/2024] [Accepted: 03/17/2024] [Indexed: 04/09/2024]
Abstract
OBJECTIVES Trace amines are powerful neuromodulators influencing the release and reuptake of catecholamines. These low concentrated endogenous amines impact mood, cognition, and hormone regulation. Dysregulation of trace amines have been associated with a variety of diseases, such as schizophrenia, Parkinson's disease, migraine, depression and more. Succesfull simultaneous quantification of trace amines, their precursors and metabolites would benefit both research and patient care. Since these compounds have various functional groups and are present in biological matrices with large concentration difference, their simultaneous quantification is an analytical challenge. Our goal was to develop a highly sensitive LC-MS/MS assay to simultaneously quantify trace amines, their precursors and metabolites in plasma. METHODS Our method is based on a simple two-step in-matrix derivatization protocol: propionic anhydride (PA) and 3-Ethyl-1-[3-(dimethylamino)propyl]carbodiimide (EDC) in combination with 2,2,2-trifluoroethylamine (TFEA) followed by online solid phase extraction combined with LC-MS/MS. Fifteen metabolites can be measured simultaneously, three precursors, eight trace amines and four metabolites. Validation of this method was performed according to international validation guidelines. The pre-analytical stability of trace amines was assessed. RESULTS This novel method was successful in quantifying trace amines, their precursors, and metabolites in plasma. Using just 50 µl human plasma, we were able to accomplish limit of quantification for 2-phenylethylamine and N-methyl-phenylethylamine of 0.2 nmol/L and 0.1 nmol/L for tyramine and n-methyltyramine. Inter-and intra-assay imprecision was < 15 % for all analytes. Stability assessment showed susceptibility of certain trace amines e.g. 2-phenylethylamine and N-methyl-phenylethylamine to enzymatic degradation in plasma. The addition of the monoamine oxidase inhibitor pargyline to plasma prevented this enzymatic degradation. CONCLUSIONS We developed a novel LC-MS/MS method that1) uses a new double derivatization technique, 2) is automated with online SPE, 3) uses far less sample volume then previous methods and 4) detects more components in the same sample (eight trace amines, three precursors, and four metabolites) with high specificity and selectivity. Furthermore, addition of MAO A/B inhibitor prevents degradation and guarantees more accurate quantification of trace amines.
Collapse
Affiliation(s)
- Krisztina de Bruyn
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, 9700 RB, The Netherlands
| | - Eugene F Diekman
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, 9700 RB, The Netherlands
| | - Claude P van der Ley
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, 9700 RB, The Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, 9700 RB, The Netherlands.
| | - Ido P Kema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, 9700 RB, The Netherlands
| |
Collapse
|
15
|
Beydoun HA, Beydoun MA, Wassertheil-Smoller S, Saquib N, Manson JE, Snetselaar L, Weiss J, Zonderman AB, Brunner R. Depressive symptoms and antidepressant use in relation to white blood cell count among postmenopausal women from the Women's Health Initiative. Transl Psychiatry 2024; 14:157. [PMID: 38514652 PMCID: PMC10958010 DOI: 10.1038/s41398-024-02872-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 03/23/2024] Open
Abstract
Inflammation can play a role in the pathophysiology of depression, and specific types of antidepressants may have inflammatory or anti-inflammatory properties. Furthermore, depression and antidepressant use has been linked to white blood cell (WBC) count, a routinely measured inflammatory marker. We examined the cross-sectional and longitudinal relationships of depressive symptoms and/or antidepressant use with WBC count among postmenopausal women. Analyses of cross-sectional data at enrollment were performed on 125,307 participants, 50-79 years of age, from the Women's Health Initiative Clinical Trials and Observational Studies who met eligibility criteria, and a subset of those with 3-year follow-up data were examined for longitudinal relationships. Depressive symptoms were defined using the Burnam Algorithm whereas antidepressant use was defined using therapeutic class codes. WBC count (Kcell/ml) was obtained through laboratory evaluations of fasting blood samples. Multivariable regression modeling was performed taking sociodemographic, lifestyle and health characteristics into consideration. At enrollment, nearly 85% were non-users of antidepressants with no depressive symptoms, 5% were antidepressant users with no depressive symptoms, 9% were non-users of antidepressants with depressive symptoms, and 2% were users of antidepressants with depressive symptoms. In fully-adjusted models, cross-sectional relationships were observed whereby women in the 2nd (OR = 1.06, 95% CI: 1.01, 1.13), 3rd (OR = 1.06, 95% CI: 1.00, 1.12) or 4th (OR = 1.10, 95% CI: 1.05, 1.17) quartiles of WBC count were more likely to exhibit depressive symptoms, and women in the 4th quartile were more likely to be users of antidepressants (OR = 1.07, 95% CI: 1.00, 1.15), compared to women in the 1st quartile. Compared to women who exhibited no depressive symptoms at either visit, those with consistent depressive symptoms at enrollment and at 3-year follow-up had faster decline in WBC count (β = -0.73, 95% CI: -1.33, -0.14) over time. No significant bidirectional relationships were observed between changes in depressive symptoms score and WBC count over time. In conclusion, depressive symptoms and/or antidepressant use were cross-sectionally related to higher WBC counts among postmenopausal women. Further evaluation of observed relationships is needed in the context of prospective cohort studies involving older adult men and women, with repeated measures of depression, antidepressant use, and WBC count.
Collapse
Affiliation(s)
- Hind A Beydoun
- Department of Research Programs, Fort Belvoir Community Hospital, Fort Belvoir, VA, USA.
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, USA.
| | - May A Beydoun
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, USA
| | | | - Nazmus Saquib
- College of Medicine, Sulaiman AlRajhi University, Al Bukairiyah, Kingdom of Saudi Arabia
| | - JoAnn E Manson
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Linda Snetselaar
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, USA
| | - Jordan Weiss
- Department of Demography, UC Berkeley, Berkeley, CA, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIA/NIH/IRP, Baltimore, MD, USA
| | - Robert Brunner
- Department of Family and Community Medicine (Emeritus), School of Medicine, University of Nevada (Reno), Reno, NV, USA
| |
Collapse
|
16
|
Abstract
Psychotropic drug-related weight gain (PDWG) is a common occurrence and is highly associated with non-initiation, discontinuation, and dissatisfaction with psychiatric drugs. Moreover, PDWG intersects with the elevated risk for obesity and associated morbidity that has been amply reported in the psychiatric population. Evidence indicates that differential liability for PDWG exists for antipsychotics, antidepressants, and anticonvulsants. During the past two decades, agents within these classes have become available with significantly lower or no liability for PDWG and as such should be prioritized. Although lithium is associated with weight gain, the overall extent of weight gain is significantly lower than previously estimated. The benefit of lifestyle and behavioral modification for obesity and/or PDWG in psychiatric populations is established, with effectiveness similar to that in the general population. Metformin is the most studied pharmacological treatment in the prevention and treatment of PDWG, and promising data are emerging for glucagon-like peptide-1 (GLP-1) receptor agonists (e.g., liraglutide, exenatide, semaglutide). Most pharmacologic antidotes for PDWG are supported with low-confidence data (e.g., topiramate, histamine-2 receptor antagonists). Future vistas for pharmacologic treatment for PDWG include large, adequately controlled studies with GLP-1 receptor agonists and possibly GLP-1/glucose-dependent insulinotropic polypeptide co-agonists (e.g., tirzepatide) as well as specific dietary modifications.
Collapse
Affiliation(s)
- Roger S McIntyre
- Department of Psychiatry (McIntyre, Rosenblat, Mansur) and Department of Pharmacology and Toxicology (McIntyre, Rosenblat, Mansur), University of Toronto, Toronto; Brain and Cognition Discovery Foundation, Toronto (McIntyre, Kwan, Teopiz); Faculty of Medicine, University of Ottawa, Ottawa (Kwan)
| | - Angela T H Kwan
- Department of Psychiatry (McIntyre, Rosenblat, Mansur) and Department of Pharmacology and Toxicology (McIntyre, Rosenblat, Mansur), University of Toronto, Toronto; Brain and Cognition Discovery Foundation, Toronto (McIntyre, Kwan, Teopiz); Faculty of Medicine, University of Ottawa, Ottawa (Kwan)
| | - Joshua D Rosenblat
- Department of Psychiatry (McIntyre, Rosenblat, Mansur) and Department of Pharmacology and Toxicology (McIntyre, Rosenblat, Mansur), University of Toronto, Toronto; Brain and Cognition Discovery Foundation, Toronto (McIntyre, Kwan, Teopiz); Faculty of Medicine, University of Ottawa, Ottawa (Kwan)
| | - Kayla M Teopiz
- Department of Psychiatry (McIntyre, Rosenblat, Mansur) and Department of Pharmacology and Toxicology (McIntyre, Rosenblat, Mansur), University of Toronto, Toronto; Brain and Cognition Discovery Foundation, Toronto (McIntyre, Kwan, Teopiz); Faculty of Medicine, University of Ottawa, Ottawa (Kwan)
| | - Rodrigo B Mansur
- Department of Psychiatry (McIntyre, Rosenblat, Mansur) and Department of Pharmacology and Toxicology (McIntyre, Rosenblat, Mansur), University of Toronto, Toronto; Brain and Cognition Discovery Foundation, Toronto (McIntyre, Kwan, Teopiz); Faculty of Medicine, University of Ottawa, Ottawa (Kwan)
| |
Collapse
|
17
|
Liu H, Zheng Y, Wang Y, Wang Y, He X, Xu P, Huang S, Yuan Q, Zhang X, Wang L, Jiang K, Chen H, Li Z, Liu W, Wang S, Xu HE, Xu F. Recognition of methamphetamine and other amines by trace amine receptor TAAR1. Nature 2023; 624:663-671. [PMID: 37935377 DOI: 10.1038/s41586-023-06775-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/20/2023] [Indexed: 11/09/2023]
Abstract
Trace amine-associated receptor 1 (TAAR1), the founding member of a nine-member family of trace amine receptors, is responsible for recognizing a range of biogenic amines in the brain, including the endogenous β-phenylethylamine (β-PEA)1 as well as methamphetamine2, an abused substance that has posed a severe threat to human health and society3. Given its unique physiological role in the brain, TAAR1 is also an emerging target for a range of neurological disorders including schizophrenia, depression and drug addiction2,4,5. Here we report structures of human TAAR1-G-protein complexes bound to methamphetamine and β-PEA as well as complexes bound to RO5256390, a TAAR1-selective agonist, and SEP-363856, a clinical-stage dual agonist for TAAR1 and serotonin receptor 5-HT1AR (refs. 6,7). Together with systematic mutagenesis and functional studies, the structures reveal the molecular basis of methamphetamine recognition and underlying mechanisms of ligand selectivity and polypharmacology between TAAR1 and other monoamine receptors. We identify a lid-like extracellular loop 2 helix/loop structure and a hydrogen-bonding network in the ligand-binding pockets, which may contribute to the ligand recognition in TAAR1. These findings shed light on the ligand recognition mode and activation mechanism for TAAR1 and should guide the development of next-generation therapeutics for drug addiction and various neurological disorders.
Collapse
Affiliation(s)
- Heng Liu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - You Zheng
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yue Wang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yumeng Wang
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecule Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Xinheng He
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Peiyu Xu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sijie Huang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qingning Yuan
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- The Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xinyue Zhang
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ling Wang
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kexin Jiang
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hong Chen
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai, China
- Shanghai Yuansi Standard Science and Technology Co., Ltd, Shanghai, China
| | - Zhen Li
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenbin Liu
- Shanghai Key Laboratory of Crime Scene Evidence, Shanghai Research Institute of Criminal Science and Technology, Shanghai, China.
- Shanghai Yuansi Standard Science and Technology Co., Ltd, Shanghai, China.
| | - Sheng Wang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecule Cell Science, Chinese Academy of Sciences, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| | - H Eric Xu
- The State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Fei Xu
- iHuman Institute, School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
| |
Collapse
|
18
|
Cichero E, Francesconi V, Casini B, Casale M, Kanov E, Gerasimov AS, Sukhanov I, Savchenko A, Espinoza S, Gainetdinov RR, Tonelli M. Discovery of Guanfacine as a Novel TAAR1 Agonist: A Combination Strategy through Molecular Modeling Studies and Biological Assays. Pharmaceuticals (Basel) 2023; 16:1632. [PMID: 38004497 PMCID: PMC10674299 DOI: 10.3390/ph16111632] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Trace amine-associated receptor 1 (TAAR1) is an attractive target for the design of innovative drugs to be applied in diverse pharmacological settings. Due to a non-negligible structural similarity with endogenous ligands, most of the agonists developed so far resulted in being affected by a low selectivity for TAAR1 with respect to other monoaminergic G protein-coupled receptors, like the adrenoreceptors. This study utilized comparative molecular docking studies and quantitative-structure activity relationship (QSAR) analyses to unveil key structural differences between TAAR1 and alpha2-adrenoreceptor (α2-ADR), with the aim to design novel TAAR1 agonists characterized by a higher selectivity profile and reduced off-target effects. While the presence of hydrophobic motives is encouraged towards both the two receptors, the introduction of polar/positively charged groups and the ligand conformation deeply affect the TAAR1 or α2-ADR putative selectivity. These computational methods allowed the identification of the α2A-ADR agonist guanfacine as an attractive TAAR1-targeting lead compound, demonstrating nanomolar activity in vitro. In vivo exploration of the efficacy of guanfacine showed that it is able to decrease the locomotor activity of dopamine transporter knockout (DAT-KO) rats. Therefore, guanfacine can be considered as an interesting template molecule worthy of structural optimization. The dual activity of guanfacine on both α2-ADR and TAAR1 signaling and the related crosstalk between the two pathways will deserve more in-depth investigation.
Collapse
Affiliation(s)
- Elena Cichero
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, 16132 Genoa, Italy; (E.C.); (V.F.); (B.C.)
| | - Valeria Francesconi
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, 16132 Genoa, Italy; (E.C.); (V.F.); (B.C.)
| | - Beatrice Casini
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, 16132 Genoa, Italy; (E.C.); (V.F.); (B.C.)
| | - Monica Casale
- Section of Chemistry and Food and Pharmaceutical Technologies, University of Genoa, 16148 Genoa, Italy;
| | - Evgeny Kanov
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (E.K.); (A.S.G.); (R.R.G.)
- St. Petersburg University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Andrey S. Gerasimov
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (E.K.); (A.S.G.); (R.R.G.)
| | - Ilya Sukhanov
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, 197022 St. Petersburg, Russia; (I.S.); (A.S.)
| | - Artem Savchenko
- Valdman Institute of Pharmacology, Pavlov First St. Petersburg State Medical University, 197022 St. Petersburg, Russia; (I.S.); (A.S.)
| | - Stefano Espinoza
- Department of Health Sciences and Research Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), 28100 Novara, Italy;
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), 16152 Genova, Italy
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, 199034 St. Petersburg, Russia; (E.K.); (A.S.G.); (R.R.G.)
- St. Petersburg University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Michele Tonelli
- Department of Pharmacy, Section of Medicinal Chemistry, School of Medical and Pharmaceutical Sciences, University of Genoa, 16132 Genoa, Italy; (E.C.); (V.F.); (B.C.)
| |
Collapse
|
19
|
Shabani S, Houlton S, Ghimire B, Tonello D, Reed C, Baba H, Aldrich S, Phillips TJ. Robust aversive effects of trace amine-associated receptor 1 activation in mice. Neuropsychopharmacology 2023; 48:1446-1454. [PMID: 37055488 PMCID: PMC10425385 DOI: 10.1038/s41386-023-01578-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/03/2023] [Accepted: 03/26/2023] [Indexed: 04/15/2023]
Abstract
Drugs that stimulate the trace amine-associated receptor 1 (TAAR1) are under clinical investigation as treatments for several neuropsychiatric disorders. Previous studies in a genetic mouse model of voluntary methamphetamine intake identified TAAR1, expressed by the Taar1 gene, as a critical mediator of aversive methamphetamine effects. Methamphetamine is a TAAR1 agonist, but also has actions at monoamine transporters. Whether exclusive activation of TAAR1 has aversive effects was not known at the time we conducted our studies. Mice were tested for aversive effects of the selective TAAR1 agonist, RO5256390, using taste and place conditioning procedures. Hypothermic and locomotor effects were also examined, based on prior evidence of TAAR1 mediation. Male and female mice of several genetic models were used, including lines selectively bred for high and low methamphetamine drinking, a knock-in line in which a mutant form of Taar1 that codes for a non-functional TAAR1 was replaced by the reference Taar1 allele that codes for functional TAAR1, and their matched control line. RO5256390 had robust aversive, hypothermic and locomotor suppressing effects that were found only in mice with functional TAAR1. Knock-in of the reference Taar1 allele rescued these phenotypes in a genetic model that normally lacks TAAR1 function. Our study provides important data on TAAR1 function in aversive, locomotor, and thermoregulatory effects that are important to consider when developing TAAR1 agonists as therapeutic drugs. Because other drugs can have similar consequences, potential additive effects should be carefully considered as these treatment agents are being developed.
Collapse
Affiliation(s)
- Shkelzen Shabani
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, USA
- Department of Biology, Minot State University, Minot, ND, USA
- Biomedical Sciences at Grand Valley State University, Allendale, MI, USA
| | - Sydney Houlton
- Department of Biology, Minot State University, Minot, ND, USA
| | - Bikalpa Ghimire
- Department of Biology, Minot State University, Minot, ND, USA
| | - Derek Tonello
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, USA
| | - Cheryl Reed
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Harue Baba
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Sara Aldrich
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Tamara J Phillips
- Department of Behavioral Neuroscience and Methamphetamine Abuse Research Center, Oregon Health & Science University, Portland, OR, USA.
- VA Portland Health Care System, Portland, OR, USA.
| |
Collapse
|
20
|
Le GH, Gillissie ES, Rhee TG, Cao B, Alnefeesi Y, Guo Z, Di Vincenzo JD, Jawad MY, March AM, Ramachandra R, Lui LMW, McIntyre RS. Efficacy, safety, and tolerability of ulotaront (SEP-363856, a trace amine-associated receptor 1 agonist) for the treatment of schizophrenia and other mental disorders with similar pathophysiology: a systematic review of preclinical and clinical trials. Expert Opin Investig Drugs 2023:1-15. [PMID: 37096491 DOI: 10.1080/13543784.2023.2206559] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
INTRODUCTION Schizophrenia is a mental illness that can disrupt emotions, perceptions, cognition, and reduce quality of life. The classical approach to treat schizophrenia uses typical and atypical antipsychotics; however, limitations include low efficacy in mitigating negative symptoms and cognitive dysfunctions, and a range of adverse effects. Evidence has accumulated on trace amine-associated receptor 1 (TAAR1) as a novel therapeutic target for treating schizophrenia. This systematic review investigates the available evidence on a TAAR1 agonist, ulotaront, as a treatment for schizophrenia. METHODS A systematic search was conducted on PubMed/MEDLINE, and Ovid databases for English-published articles from inception to December 18, 2022. Literature focusing on the association between ulotaront and schizophrenia were evaluated based on an inclusion/exclusion criterion. Selected studies were assessed for risk of bias, using Cochrane Collaboration tool, and summarized in a table to generate discussion topics. RESULTS Three clinical, two comparative, and five preclinical studies examining ulotaront's pharmacology, tolerability and safety, and/or efficacy were identified. Results indicate that ulotaront has a differing adverse effects profile from other antipsychotics, may mitigate metabolic-related adverse effects commonly associated with antipsychotics, and may be effective for treating positive and negative symptoms. CONCLUSIONS Findings from available literature present ulotaront as a potential and promising alternative treatment method for schizophrenia. Despite this, our results were limited due to lack of clinical trials on ulotaront's long-term efficacy and mechanisms of action. Future research should focus on these limitations to elucidate ulotaront's efficacy and safety for the treatment of schizophrenia and other mental disorders with similar pathophysiology.
Collapse
Affiliation(s)
- Gia Han Le
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Emily S Gillissie
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Taeho Greg Rhee
- Department of Psychiatry, School of Medicine, Yale University, New Haven, CT, USA
- VA New England Mental Illness, Research, Education and Clinical Center (MIRECC), VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Public Health Sciences, School of Medicine, University of Connecticut, Farmington, CT, USA
| | - Bing Cao
- Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, 400715, P. R. China
| | - Yazen Alnefeesi
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Ziji Guo
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Joshua D Di Vincenzo
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Muhammad Youshay Jawad
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Andrew M March
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
| | - Ranuk Ramachandra
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Leanna M W Lui
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, ON, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
- Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| |
Collapse
|
21
|
Ovrom EA, Mostert KA, Khakhkhar S, McKee DP, Yang P, Her YF. A Comprehensive Review of the Genetic and Epigenetic Contributions to the Development of Fibromyalgia. Biomedicines 2023; 11:1119. [PMID: 37189737 PMCID: PMC10135661 DOI: 10.3390/biomedicines11041119] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 05/17/2023] Open
Abstract
This narrative review summarizes the current knowledge of the genetic and epigenetic contributions to the development of fibromyalgia (FM). Although there is no single gene that results in the development of FM, this study reveals that certain polymorphisms in genes involved in the catecholaminergic pathway, the serotonergic pathway, pain processing, oxidative stress, and inflammation may influence susceptibility to FM and the severity of its symptoms. Furthermore, epigenetic changes at the DNA level may lead to the development of FM. Likewise, microRNAs may impact the expression of certain proteins that lead to the worsening of FM-associated symptoms.
Collapse
Affiliation(s)
- Erik A. Ovrom
- Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA;
| | - Karson A. Mostert
- Department of Physical Medicine and Rehabilitation, Mayo Clinic Hospital, Rochester, MN 55905, USA
| | - Shivani Khakhkhar
- Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Daniel P. McKee
- Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Padao Yang
- Department of Psychiatry and Psychology, Mayo Clinic Hospital, Rochester, MN 55905, USA
| | - Yeng F. Her
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic Hospital, Rochester, MN 55905, USA
| |
Collapse
|
22
|
McIntyre RS. Rapid-acting antidepressants in psychiatry: episodic treatments, innovation, and clarion call for methodologic rigor in drug development. Expert Opin Drug Saf 2022; 21:715-716. [PMID: 35818802 DOI: 10.1080/14740338.2022.2100595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Roger S McIntyre
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| |
Collapse
|
23
|
Alnefeesi Y, Chen-Li D, Krane E, Jawad MY, Rodrigues NB, Ceban F, Di Vincenzo JD, Meshkat S, Ho RCM, Gill H, Teopiz KM, Cao B, Lee Y, McIntyre RS, Rosenblat JD. Real-world effectiveness of ketamine in treatment-resistant depression: A systematic review & meta-analysis. J Psychiatr Res 2022; 151:693-709. [PMID: 35688035 DOI: 10.1016/j.jpsychires.2022.04.037] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/10/2022] [Accepted: 04/26/2022] [Indexed: 12/29/2022]
Abstract
Ketamine is a promising therapeutic option in treatment-resistant depression (TRD). The acute efficacy of ketamine in TRD has been demonstrated in replicated randomised-controlled trials (RCTs), but the generalizability of RCT data to real-world practice is limited. To this end, we conducted a systematic review (Search date: 25/12/2021; 1482 records identified) and meta-analysis of studies evaluating the real-world clinical effectiveness of ketamine in TRD patients. Four overlapping syntheses (Total n = 2665 patients; k = 79 studies) and 32 meta-regressions (Total n = 2050; k = 37) were conducted. All results suggest that the mean antidepressant effect is substantial (mean ± 95% CI, % responded = 45 ± 10%; p< 0.0001, % remitted = 30 ± 5.9%; p< 0.0001, Hedges g of symptomatological improvement = 1.44 ± 0.609; p < 0.0001), but the effect varies considerably among patients. The more treatment-resistant cases were found to remit less often (p < 0.01), but no such effect on response was evident (p > 0.05). Meta-regressions also confirmed that the therapeutic effect does not significantly decline with repeated treatments (p > 0.05). These results demonstrate that even the most treatment-resistant patients may benefit from ketamine, and that mid-to-long term treatment is effective in many patients.
Collapse
Affiliation(s)
- Yazen Alnefeesi
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - David Chen-Li
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Ella Krane
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | | | - Nelson B Rodrigues
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Felicia Ceban
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Joshua D Di Vincenzo
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Shakila Meshkat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Roger C M Ho
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute of Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore
| | - Hartej Gill
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Kayla M Teopiz
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Bing Cao
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Key Laboratory of Cognition and Personality, Faculty of Psychology, Ministry of Education, Southwest University, Chongqing, 400715, PR China
| | - Yena Lee
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada
| | - Roger S McIntyre
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Department of Pharmacology, University of Toronto, Toronto, ON, Canada; Brain and Cognition Discovery Foundation, Toronto, ON, Canada
| | - Joshua D Rosenblat
- Mood Disorders Psychopharmacology Unit, University Health Network, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|