1
|
Ubhayarathna M, Langmead CJ, Diepenhorst NA, Stewart GD. Molecular and structural insights into the 5-HT 2C receptor as a therapeutic target for substance use disorders. Br J Pharmacol 2024; 181:4414-4429. [PMID: 37679998 DOI: 10.1111/bph.16233] [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: 11/03/2022] [Revised: 08/07/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023] Open
Abstract
Substance use disorder (SUD) is a chronic condition, with maintained abuse of a substance leading to physiological and psychological alterations and often changes in cognitive and social behaviours. Current therapies include psychotherapy coupled with medication; however, high relapse rates reveal the shortcomings of these therapies. The signalling, expression profile, and neurological function of the serotonin 2C receptor (5-HT2C receptor) make it a candidate of interest for the treatment of SUD. Recently, psychedelics, which broadly act at 5-HT2 receptors, have indicated potential for the treatment of SUD, implicating the 5-HT2C receptor. The modern psychedelic movement has rekindled interest in the 5-HT2C receptor, resulting in many new studies, especially structural analyses. This review explores the structural, molecular and cellular mechanisms governing 5-HT2C receptor function in the context of SUD. This provides the basis of the preclinical and clinical evidence for their role in SUD and highlights the potential for future exploration.
Collapse
Affiliation(s)
- Maleesha Ubhayarathna
- Drug Discovery Biology and Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Christopher J Langmead
- Drug Discovery Biology and Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Australia
| | - Natalie A Diepenhorst
- Drug Discovery Biology and Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Gregory D Stewart
- Drug Discovery Biology and Neuroscience & Mental Health Therapeutic Program Area, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Australia
| |
Collapse
|
2
|
Sandoval KE, Witt KA. Somatostatin: Linking Cognition and Alzheimer Disease to Therapeutic Targeting. Pharmacol Rev 2024; 76:1291-1325. [PMID: 39013601 PMCID: PMC11549939 DOI: 10.1124/pharmrev.124.001117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
Over 4 decades of research support the link between Alzheimer disease (AD) and somatostatin [somatotropin-releasing inhibitory factor (SRIF)]. SRIF and SRIF-expressing neurons play an essential role in brain function, modulating hippocampal activity and memory formation. Loss of SRIF and SRIF-expressing neurons in the brain rests at the center of a series of interdependent pathological events driven by amyloid-β peptide (Aβ), culminating in cognitive decline and dementia. The connection between the SRIF and AD further extends to the neuropsychiatric symptoms, seizure activity, and inflammation, whereas preclinical AD investigations show SRIF or SRIF receptor agonist administration capable of enhancing cognition. SRIF receptor subtype-4 activation in particular presents unique attributes, with the potential to mitigate learning and memory decline, reduce comorbid symptoms, and enhance enzymatic degradation of Aβ in the brain. Here, we review the links between SRIF and AD along with the therapeutic implications. SIGNIFICANCE STATEMENT: Somatostatin and somatostatin-expressing neurons in the brain are extensively involved in cognition. Loss of somatostatin and somatostatin-expressing neurons in Alzheimer disease rests at the center of a series of interdependent pathological events contributing to cognitive decline and dementia. Targeting somatostatin-mediated processes has significant therapeutic potential for the treatment of Alzheimer disease.
Collapse
Affiliation(s)
- Karin E Sandoval
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| | - Ken A Witt
- Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, Illinois
| |
Collapse
|
3
|
Cui L, Li S, Wang S, Wu X, Liu Y, Yu W, Wang Y, Tang Y, Xia M, Li B. Major depressive disorder: hypothesis, mechanism, prevention and treatment. Signal Transduct Target Ther 2024; 9:30. [PMID: 38331979 PMCID: PMC10853571 DOI: 10.1038/s41392-024-01738-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/24/2023] [Accepted: 12/28/2023] [Indexed: 02/10/2024] Open
Abstract
Worldwide, the incidence of major depressive disorder (MDD) is increasing annually, resulting in greater economic and social burdens. Moreover, the pathological mechanisms of MDD and the mechanisms underlying the effects of pharmacological treatments for MDD are complex and unclear, and additional diagnostic and therapeutic strategies for MDD still are needed. The currently widely accepted theories of MDD pathogenesis include the neurotransmitter and receptor hypothesis, hypothalamic-pituitary-adrenal (HPA) axis hypothesis, cytokine hypothesis, neuroplasticity hypothesis and systemic influence hypothesis, but these hypothesis cannot completely explain the pathological mechanism of MDD. Even it is still hard to adopt only one hypothesis to completely reveal the pathogenesis of MDD, thus in recent years, great progress has been made in elucidating the roles of multiple organ interactions in the pathogenesis MDD and identifying novel therapeutic approaches and multitarget modulatory strategies, further revealing the disease features of MDD. Furthermore, some newly discovered potential pharmacological targets and newly studied antidepressants have attracted widespread attention, some reagents have even been approved for clinical treatment and some novel therapeutic methods such as phototherapy and acupuncture have been discovered to have effective improvement for the depressive symptoms. In this work, we comprehensively summarize the latest research on the pathogenesis and diagnosis of MDD, preventive approaches and therapeutic medicines, as well as the related clinical trials.
Collapse
Affiliation(s)
- Lulu Cui
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Shu Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Siman Wang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Xiafang Wu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Yingyu Liu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Weiyang Yu
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Yijun Wang
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China
- China Medical University Centre of Forensic Investigation, Shenyang, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling/Key Laboratory of Acupuncture for Senile Disease (Chengdu University of TCM), Ministry of Education/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine/Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Maosheng Xia
- Department of Orthopaedics, The First Hospital, China Medical University, Shenyang, China.
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, China.
- China Medical University Centre of Forensic Investigation, Shenyang, China.
| |
Collapse
|
4
|
Mendez-David I, David DJ, Deloménie C, Tritschler L, Beaulieu JM, Colle R, Corruble E, Gardier AM, Hen R. A complex relation between levels of adult hippocampal neurogenesis and expression of the immature neuron marker doublecortin. Hippocampus 2023; 33:1075-1093. [PMID: 37421207 DOI: 10.1002/hipo.23568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 05/08/2023] [Accepted: 06/20/2023] [Indexed: 07/10/2023]
Abstract
We investigated the mechanisms underlying the effects of the antidepressant fluoxetine on behavior and adult hippocampal neurogenesis (AHN). After confirming our earlier report that the signaling molecule β-arrestin-2 (β-Arr2) is required for the antidepressant-like effects of fluoxetine, we found that the effects of fluoxetine on proliferation of neural progenitors and survival of adult-born granule cells are absent in the β-Arr2 knockout (KO) mice. To our surprise, fluoxetine induced a dramatic upregulation of the number of doublecortin (DCX)-expressing cells in the β-Arr2 KO mice, indicating that this marker can be increased even though AHN is not. We discovered two other conditions where a complex relationship occurs between the number of DCX-expressing cells compared to levels of AHN: a chronic antidepressant model where DCX is upregulated and an inflammation model where DCX is downregulated. We concluded that assessing the number of DCX-expressing cells alone to quantify levels of AHN can be complex and that caution should be applied when label retention techniques are unavailable.
Collapse
Affiliation(s)
- Indira Mendez-David
- Université Paris-Saclay, UVSQ, Centre de recherche en Epidémiologie et Santé des Populations (CESP), UMR 1018, CESP-Inserm, Team Moods, Faculté de Pharmacie, Bâtiment Henri MOISSAN, Orsay, France
- Department of Psychiatry, Columbia University, New York, New York, USA
- Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York, USA
| | - Denis Joseph David
- Université Paris-Saclay, UVSQ, Centre de recherche en Epidémiologie et Santé des Populations (CESP), UMR 1018, CESP-Inserm, Team Moods, Faculté de Pharmacie, Bâtiment Henri MOISSAN, Orsay, France
- Department of Psychiatry, Columbia University, New York, New York, USA
- Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York, USA
| | - Claudine Deloménie
- UMS-IPSIT ACTAGen, Inserm, CNRS, Ingénierie et Plateformes au Service de l'Innovation Thérapeutique, Université Paris-Saclay, Bâtiment Henri MOISSAN, Orsay, France
| | - Laurent Tritschler
- Université Paris-Saclay, UVSQ, Centre de recherche en Epidémiologie et Santé des Populations (CESP), UMR 1018, CESP-Inserm, Team Moods, Faculté de Pharmacie, Bâtiment Henri MOISSAN, Orsay, France
| | - Jean-Martin Beaulieu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Romain Colle
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Emmanuelle Corruble
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, Univ Paris-Saclay, Le Kremlin Bicêtre, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Alain Michel Gardier
- Université Paris-Saclay, UVSQ, Centre de recherche en Epidémiologie et Santé des Populations (CESP), UMR 1018, CESP-Inserm, Team Moods, Faculté de Pharmacie, Bâtiment Henri MOISSAN, Orsay, France
| | - René Hen
- Department of Psychiatry, Columbia University, New York, New York, USA
- Division of Integrative Neuroscience, New York State Psychiatric Institute, New York, New York, USA
| |
Collapse
|
5
|
Kim C, Lee DY, Park J, Yang SJ, Tan EH, Prieto-Alhambra D, Lee YH, Lee S, Kim SJ, Lee J, Park RW, Shin Y. Safety outcomes of selective serotonin reuptake inhibitors in adolescent attention-deficit/hyperactivity disorder with comorbid depression: the ASSURE study. Psychol Med 2023; 53:4811-4819. [PMID: 36803587 DOI: 10.1017/s0033291723000120] [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] [Indexed: 02/22/2023]
Abstract
BACKGROUND Attention deficit-hyperactivity disorder (ADHD) is related to depressive disorder, and adolescents with both present poor outcomes. However, evidence for the safety of concomitantly using a methylphenidate (MPH) and a selective serotonin reuptake inhibitor (SSRI) among adolescent ADHD patients is limited, a literature gap aimed to address through this investigation. METHODS We conducted a new-user cohort study using a nationwide claims database in South Korea. We identified a study population as adolescents who were diagnosed both ADHD and depressive disorder. MPH-only users were compared with patients who prescribed both a SSRI and a MPH. Fluoxetine and escitalopram users were also compared to find a preferable treatment option. Thirteen outcomes including neuropsychiatric, gastrointestinal, and other events were assessed, taking respiratory tract infection as a negative control outcome. We matched the study groups using a propensity score and used the Cox proportional hazard model to calculate the hazard ratio. Subgroup and sensitivity analyses were conducted in various epidemiologic settings. RESULTS The risks of all the outcomes between the MPH-only and SSRI groups were not significantly different. Regarding SSRI ingredients, the risk of tic disorder was significantly lower in the fluoxetine group than the escitalopram group [HR 0.43 (0.25-0.71)]. However, there was no significant difference in other outcomes between the fluoxetine and escitalopram groups. CONCLUSION The concomitant use of MPHs and SSRIs showed generally safe profiles in adolescent ADHD patients with depression. Most of the differences between fluoxetine and escitalopram, except those concerning tic disorder, were not significant.
Collapse
Affiliation(s)
- Chungsoo Kim
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
| | - Dong Yun Lee
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, South Korea
| | - Jimyung Park
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
| | - Su-Jin Yang
- Gwangju Smile Center for Crime victim support, Gwangju, South Korea
| | - Eng Hooi Tan
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Centre of Statistics in Medicines, University of Oxford, Oxford, UK
| | - Daniel Prieto-Alhambra
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, Centre of Statistics in Medicines, University of Oxford, Oxford, UK
- Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Yo Han Lee
- Department of Preventive Medicine and Public Health, Ajou University School of Medicine, Suwon, South Korea
| | - Sangha Lee
- Department of Psychiatry, Ajou University School of Medicine, Suwon, South Korea
| | - Seong-Ju Kim
- Department of Psychiatry, Ajou University School of Medicine, Suwon, South Korea
| | - Jeewon Lee
- Department of Psychiatry, Soonchunhyang University Bucheon Hospital, Bucheon, South Korea
| | - Rae Woong Park
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, South Korea
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, South Korea
| | - Yunmi Shin
- Department of Psychiatry, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
6
|
Presa MH, Rocha MJD, Pires CS, Ledebuhr KNB, Costa GPD, Alves D, Bortolatto CF, Brüning CA. Antidepressant-like Effect of 1-(2-(4-(4-Ethylphenyl)-1 H-1,2,3-triazol-1-yl)phenyl)ethan-1-one in Mice: Evidence of the Contribution of the Serotonergic System. ACS Chem Neurosci 2023. [PMID: 37294690 DOI: 10.1021/acschemneuro.3c00108] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023] Open
Abstract
Major depressive disorder (MDD) is a psychiatric disorder that affects a large portion of the population, with dysregulation of the serotonergic system, which is deeply involved in both the pathophysiology of MDD and mechanism of action of many antidepressants. Current pharmacological therapies do not meet the neurobiological needs of all depressed individuals, making the development of new antidepressants necessary. In recent decades, compounds containing triazoles have become promising due to their range of biological activities, including antidepressant activity. In this study, we evaluated the antidepressant-like effect of a hybrid containing triazole and acetophenone, 1-(2-(4-(4-ethylphenyl)-1H-1,2,3-triazol-1-yl)phenyl)ethan-1-one (ETAP) (0.5-5 mg/kg), in the forced swimming test (FST) and tail suspension test (TST) in mice, as well as the involvement of the serotonergic system in this effect. Our findings demonstrated that ETAP exhibited an antidepressant-like effect from the dose of 1 mg/kg and that this effect is modulated by 5-HT2A/2C and 5-HT4 receptors. We also demonstrated that this effect may be related to inhibition of monoamine oxidase A activity in the hippocampus. Additionally, we evaluated the in silico pharmacokinetic profile of ETAP, which predicted its penetration into the central nervous system. ETAP exhibited a low potential for toxicity at a high dose, making this molecule interesting for the development of a new therapeutic strategy for MDD.
Collapse
Affiliation(s)
- Marcelo Heinemann Presa
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas (UFPel), P.O. Box 354, 96010-900 Pelotas, Rio Grande do Sul, Brazil
| | - Marcia Juciele da Rocha
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas (UFPel), P.O. Box 354, 96010-900 Pelotas, Rio Grande do Sul, Brazil
| | - Camila Simões Pires
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas (UFPel), P.O. Box 354, 96010-900 Pelotas, Rio Grande do Sul, Brazil
| | - Kauane Nayara Bahr Ledebuhr
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas (UFPel), P.O. Box 354, 96010-900 Pelotas, Rio Grande do Sul, Brazil
| | - Gabriel Pereira da Costa
- Laboratory of Clean Organic Synthesis (LASOL), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas (UFPel), P.O. Box 354, 96010-900 Pelotas, Rio Grande do Sul, Brazil
| | - Diego Alves
- Laboratory of Clean Organic Synthesis (LASOL), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas (UFPel), P.O. Box 354, 96010-900 Pelotas, Rio Grande do Sul, Brazil
| | - Cristiani Folharini Bortolatto
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas (UFPel), P.O. Box 354, 96010-900 Pelotas, Rio Grande do Sul, Brazil
| | - César Augusto Brüning
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Chemical, Pharmaceutical and Food Sciences Center (CCQFA), Federal University of Pelotas (UFPel), P.O. Box 354, 96010-900 Pelotas, Rio Grande do Sul, Brazil
| |
Collapse
|
7
|
Flanigan ME, Hon OJ, D'Ambrosio S, Boyt KM, Hassanein L, Castle M, Haun HL, Pina MM, Kash TL. Subcortical serotonin 5HT 2c receptor-containing neurons sex-specifically regulate binge-like alcohol consumption, social, and arousal behaviors in mice. Nat Commun 2023; 14:1800. [PMID: 37002196 PMCID: PMC10066391 DOI: 10.1038/s41467-023-36808-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 02/17/2023] [Indexed: 04/03/2023] Open
Abstract
Binge alcohol consumption induces discrete social and arousal disturbances in human populations that promote increased drinking and accelerate the progression of Alcohol Use Disorder. Here, we show in a mouse model that binge alcohol consumption disrupts social recognition in females and potentiates sensorimotor arousal in males. These negative behavioral outcomes were associated with sex-specific adaptations in serotonergic signaling systems within the lateral habenula (LHb) and the bed nucleus of the stria terminalis (BNST), particularly those related to the receptor 5HT2c. While both BNST and LHb neurons expressing this receptor display potentiated activation following binge alcohol consumption, the primary causal mechanism underlying the effects of alcohol on social and arousal behaviors appears to be excessive activation of LHb5HT2c neurons. These findings may have valuable implications for the development of sex-specific treatments for mood and alcohol use disorders targeting the brain's serotonin system.
Collapse
Affiliation(s)
- M E Flanigan
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - O J Hon
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Curriculum in Neuroscience, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - S D'Ambrosio
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - K M Boyt
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - L Hassanein
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - M Castle
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - H L Haun
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - M M Pina
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - T L Kash
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
8
|
da Rocha MJ, Pires CS, Presa MH, Besckow EM, Nunes GD, Gomes CS, Penteado F, Lenardão EJ, Bortolatto CF, Brüning CA. Involvement of the serotonergic system in the antidepressant-like effect of 1-(phenylselanyl)-2-(p-tolyl)indolizine in mice. Psychopharmacology (Berl) 2023; 240:373-389. [PMID: 36645465 DOI: 10.1007/s00213-023-06313-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/05/2023] [Indexed: 01/17/2023]
Abstract
RATIONALE Depression is a mental disorder that affects approximately 280 million people worldwide. In the search for new treatments for mood disorders, compounds containing selenium and indolizine derivatives show promising results. OBJECTIVES AND METHODS To evaluate the antidepressant-like effect of 1-(phenylselanyl)-2-(p-tolyl)indolizine (MeSeI) (0.5-50 mg/kg, intragastric-i.g.) on the tail suspension test (TST) and the forced swim test (FST) in adult male Swiss mice and to elucidate the role of the serotonergic system in this effect through pharmacological and in silico approaches, as well to evaluate acute oral toxicity at a high dose (300 mg/kg). RESULTS MeSeI administered 30 min before the FST and the TST reduced immobility time at doses from 1 mg/kg and at 50 mg/kg and increased the latency time for the first episode of immobility, demonstrating an antidepressant-like effect. In the open field test (OFT), MeSeI did not change the locomotor activity. The antidepressant-like effect of MeSeI (50 mg/kg, i.g.) was prevented by the pre-treatment with p-chlorophenylalanine (p-CPA), a selective tryptophan hydroxylase inhibitor (100 mg/kg, intraperitoneally-i.p. for 4 days), with ketanserin, a 5-HT2A/2C receptor antagonist (1 mg/kg, i.p.), and with GR113808, a 5-HT4 receptor antagonist (0.1 mg/kg, i.p.), but not with WAY100635, a selective 5-HT1A receptor antagonist (0.1 mg/kg, subcutaneous-s.c.) and ondansetron, a 5-HT3 receptor antagonist (1 mg/kg, i.p.). MeSeI showed a binding affinity with 5-HT2A, 5 -HT2C, and 5-HT4 receptors by molecular docking. MeSeI (300 mg/kg, i.g.) demonstrated low potential to cause acute toxicity in adult female Swiss mice. CONCLUSION In summary, MeSeI exhibits an antidepressant-like effect mediated by the serotonergic system and could be considered for the development of new treatment strategies for depression.
Collapse
Affiliation(s)
- Marcia Juciele da Rocha
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Camila Simões Pires
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Marcelo Heinemann Presa
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Evelyn Mianes Besckow
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Gustavo D'Avila Nunes
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Caroline Signorini Gomes
- Clean Organic Synthesis Laboratory (LASOL), Postgraduate Program in Chemistry (PPGQ), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Filipe Penteado
- Clean Organic Synthesis Laboratory (LASOL), Postgraduate Program in Chemistry (PPGQ), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Eder João Lenardão
- Clean Organic Synthesis Laboratory (LASOL), Postgraduate Program in Chemistry (PPGQ), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil
| | - Cristiani Folharini Bortolatto
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil.
| | - César Augusto Brüning
- Laboratory of Biochemistry and Molecular Neuropharmacology (LABIONEM), Postgraduate Program in Biochemistry and Bioprospecting (PPGBBio), Center of Chemical, Pharmaceutical and Food Sciences (CCQFA), Federal University of Pelotas (UFPel), Capão Do Leão Campus, Pelotas, RS, 96010-900, Brazil.
| |
Collapse
|
9
|
Thompson SM. Plasticity of synapses and reward circuit function in the genesis and treatment of depression. Neuropsychopharmacology 2023; 48:90-103. [PMID: 36057649 PMCID: PMC9700729 DOI: 10.1038/s41386-022-01422-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/18/2022] [Accepted: 08/01/2022] [Indexed: 11/08/2022]
Abstract
What changes in brain function cause the debilitating symptoms of depression? Can we use the answers to this question to invent more effective, faster acting antidepressant drug therapies? This review provides an overview and update of the converging human and preclinical evidence supporting the hypothesis that changes in the function of excitatory synapses impair the function of the circuits they are embedded in to give rise to the pathological changes in mood, hedonic state, and thought processes that characterize depression. The review also highlights complementary human and preclinical findings that classical and novel antidepressant drugs relieve the symptoms of depression by restoring the functions of these same synapses and circuits. These findings offer a useful path forward for designing better antidepressant compounds.
Collapse
Affiliation(s)
- Scott M Thompson
- Department of Psychiatry, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, 80045, CO, USA.
| |
Collapse
|
10
|
Chronic Trazodone and Citalopram Treatments Increase Trophic Factor and Circadian Rhythm Gene Expression in Rat Brain Regions Relevant for Antidepressant Efficacy. Int J Mol Sci 2022; 23:ijms232214041. [PMID: 36430520 PMCID: PMC9698904 DOI: 10.3390/ijms232214041] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Trazodone is an efficacious atypical antidepressant acting both as an SSRI and a 5HT2A and 5HT2C antagonist. Antagonism to H1-histaminergic and alpha1-adrenergic receptors is responsible for a sleep-promoting action. We studied long-term gene expression modulations induced by chronic trazodone to investigate the molecular underpinning of trazodone efficacy. Rats received acute or chronic treatment with trazodone or citalopram. mRNA expression of growth factor and circadian rhythm genes was evaluated by qPCR in the prefrontal cortex (PFCx), hippocampus, Nucleus Accumbens (NAc), amygdala, and hypothalamus. CREB levels and phosphorylation state were evaluated using Western blotting. BDNF levels were significantly increased in PFCx and hippocampus by trazodone and in the NAc and hypothalamus by citalopram. Likewise, TrkB receptor levels augmented in the PFCx after trazodone and in the amygdala after citalopram. FGF-2 and FGFR2 levels were higher after trazodone in the PFCx. The CREB phosphorylation state was increased by chronic trazodone in the PFCx, hippocampus, and hypothalamus. Bmal1 and Per1 were increased by both antidepressants after acute and chronic treatments, while Per2 levels were specifically augmented by chronic trazodone in the PFCx and NAc, and by citalopram in the PFCx, amygdala, and NAc. These findings show that trazodone affects the expression of neurotrophic factors involved in antidepressant responses and alters circadian rhythm genes implicated in the pathophysiology of depression, thus shedding light on trazodone's molecular mechanism of action.
Collapse
|
11
|
Crawford LC, Kim S, Karelia D, Sepulveda DE, Morgan DJ, Lü J, Henderson-Redmond AN. Decursinol-mediated antinociception and anti-allodynia in acute and neuropathic pain models in male mice: Tolerance and receptor profiling. Front Pharmacol 2022; 13:968976. [PMID: 36249788 PMCID: PMC9558739 DOI: 10.3389/fphar.2022.968976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/09/2022] [Indexed: 11/26/2022] Open
Abstract
Korean scientists have shown that oral administration of Angelica gigas Nakai (AGN) root alcoholic extract and the metabolite of its pyranocoumarins, decursinol, have antinociceptive properties across various thermal and acute inflammatory pain models. The objectives of this study were 1) to assess whether tolerance develops to the antinociceptive effects of once-daily intraperitoneally administered decursinol (50 mg/kg) in acute thermal pain models, 2) to establish its anti-allodynic efficacy and potential tolerance development in a model of chemotherapy-evoked neuropathic pain (CENP) and 3) to probe the involvement of select receptors in mediating the pain-relieving effects with antagonists. The results show that decursinol induced antinociception in both the hot plate and tail-flick assays and reversed mechanical allodynia in mice with cisplatin-evoked neuropathic pain. Tolerance was detected to the antinociceptive effects of decursinol in the hot plate and tail-flick assays and to the anti-allodynic effects of decursinol in neuropathic mice. Pretreatment with either the 5-HT2 antagonist methysergide, the 5-HT2A antagonist volinanserin, or the 5-HT2C antagonist SB-242084 failed to attenuate decursinol-induced antinociception in the tail-flick assay. While pretreatment with the cannabinoid inverse agonists rimonabant and SR144528 failed to modify decursinol-induced anti-allodynia, pretreatment with the opioid antagonist naloxone partially attenuated the anti-allodynic effects of decursinol. In conclusion, our data support decursinol as an active phytochemical of AGN having both antinociceptive and anti-allodynic properties. Future work warrants a more critical investigation of potential receptor mechanisms as they are likely more complicated than initially reported.
Collapse
Affiliation(s)
- LaTaijah C. Crawford
- Biomedical Sciences Graduate Program, Pennsylvania State University College of Medicine, Hershey, PA, United States
- Department of Biomedical Sciences, Marshall University, Huntington, WV, United States
| | - Sangyub Kim
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Deepkamal Karelia
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Diana E. Sepulveda
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Daniel J. Morgan
- Department of Biomedical Sciences, Marshall University, Huntington, WV, United States
| | - Junxuan Lü
- Biomedical Sciences Graduate Program, Pennsylvania State University College of Medicine, Hershey, PA, United States
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | | |
Collapse
|
12
|
Kay LM. COVID-19 and olfactory dysfunction: a looming wave of dementia? J Neurophysiol 2022; 128:436-444. [PMID: 35894511 PMCID: PMC9377782 DOI: 10.1152/jn.00255.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
Olfactory dysfunction is a hallmark symptom of COVID-19 disease resulting from the SARS-CoV-2 virus. The cause of the sudden and usually temporary anosmia that most people suffer from COVID-19 is likely entirely peripheral-inflammation and other damage caused by the virus in the sensory epithelium inside the upper recesses of the nasal cavity can damage or prevent chemicals from properly activating the olfactory sensory neurons. However, persistent olfactory dysfunction from COVID-19, in the form of hyposmia and parosmia (decreased or altered smell) may affect as many as 15 million people worldwide. This epidemic of olfactory dysfunction is thus a continuing public health concern. Mounting evidence suggests that the SARS-CoV-2 virus itself or inflammation from the immune response in the nasal sensory epithelium may invade the olfactory bulb, likely via non-neuronal transmission. COVID-19-related long-term olfactory dysfunction and early damage to olfactory and limbic brain regions suggest a pattern of degeneration similar to that seen in early stages of Alzheimer's disease, Parkinson's disease, and Lewy body dementia. Thus, long-term olfactory dysfunction coupled with cognitive and emotional disturbance from COVID-19 may be the first signs of delayed onset dementia from neurodegeneration. Few treatments are known to be effective to prevent further degeneration, but the first line of defense against degeneration may be olfactory and environmental enrichment. There is a pressing need for more research on treatments for olfactory dysfunction and longitudinal studies including cognitive and olfactory function from patients who have recovered from even mild COVID-19.NEW & NOTEWORTHY More than 15 million people worldwide experience persistent COVID-19 olfactory dysfunction, possibly caused by olfactory bulb damage. SARS-CoV-2 can cause inflammation and viral invasion of the olfactory bulb, initiating a cascade of degeneration similar to Alzheimer's disease and Lewy body disease. People who have had even mild cases of COVID-19 show signs of degeneration in cortical areas connected with the olfactory system. These data suggest a wave of post-COVID dementia in the coming decades.
Collapse
Affiliation(s)
- Leslie M Kay
- Institute for Mind and Biology, Department of Psychology, The University of Chicago, Chicago, Illinois
| |
Collapse
|
13
|
Xu S, Yao X, Li B, Cui R, Zhu C, Wang Y, Yang W. Uncovering the Underlying Mechanisms of Ketamine as a Novel Antidepressant. Front Pharmacol 2022; 12:740996. [PMID: 35872836 PMCID: PMC9301111 DOI: 10.3389/fphar.2021.740996] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022] Open
Abstract
Major depressive disorder (MDD) is a devastating psychiatric disorder which exacts enormous personal and social-economic burdens. Ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist, has been discovered to exert rapid and sustained antidepressant-like actions on MDD patients and animal models. However, the dissociation and psychotomimetic propensities of ketamine have limited its use for psychiatric indications. Here, we review recently proposed mechanistic hypotheses regarding how ketamine exerts antidepressant-like actions. Ketamine may potentiate α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR)-mediated transmission in pyramidal neurons by disinhibition and/or blockade of spontaneous NMDAR-mediated neurotransmission. Ketamine may also activate neuroplasticity- and synaptogenesis-relevant signaling pathways, which may converge on key components like brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) and mechanistic target of rapamycin (mTOR). These processes may subsequently rebalance the excitatory/inhibitory transmission and restore neural network integrity that is compromised in depression. Understanding the mechanisms underpinning ketamine’s antidepressant-like actions at cellular and neural circuit level will drive the development of safe and effective pharmacological interventions for the treatment of MDD.
Collapse
Affiliation(s)
- Songbai Xu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Xiaoxiao Yao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Cuilin Zhu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Cuilin Zhu, ; Yao Wang, ; Wei Yang,
| | - Yao Wang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Cuilin Zhu, ; Yao Wang, ; Wei Yang,
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Cuilin Zhu, ; Yao Wang, ; Wei Yang,
| |
Collapse
|
14
|
Lewis V, Rodrigue B, Arsenault E, Zhang M, Taghavi-Abkuh FF, Silva WCC, Myers M, Matta-Camacho E, Aguilar-Valles A. Translational control by ketamine and its implications for comorbid cognitive deficits in depressive disorders. J Neurochem 2022. [PMID: 35680556 DOI: 10.1111/jnc.15652] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/19/2022] [Accepted: 05/26/2022] [Indexed: 11/29/2022]
Abstract
Ketamine has shown antidepressant effects in patients with major depressive disorder (MDD) resistant to first-line treatments and approved for use in this patient population. Ketamine induces several forms of synaptic plasticity, which are proposed to underlie its antidepressant effects. However, the molecular mechanism of action directly responsible for ketamine's antidepressant effects remains under active investigation. It was recently demonstrated that the effectors of the mammalian target of rapamycin complex 1 (mTORC1) signalling pathway, namely, eukaryotic initiation factor 4E (eIF4E) binding proteins 1 and 2 (4E-BP1 and 4E-BP2), are central in mediating ketamine-induced synaptic plasticity and behavioural antidepressant-like effect. 4E-BPs are a family of messenger ribonucleic acid (mRNA) translation repressors inactivated by mTORC1. We observed that their expression in inhibitory interneurons mediates ketamine's effects in the forced swim and novelty suppressed feeding tests and the long-lasting inhibition of GABAergic neurotransmission in the hippocampus. In addition, another effector pathway that regulates translation elongation downstream of mTORC1, the eukaryotic elongation factor 2 kinase (eEF2K), has been implicated in ketamine's behavioural effects. We will discuss how ketamine's rapid antidepressant effect depends on the activation of neuronal mRNA translation through 4E-BP1/2 and eEF2K. Furthermore, given that these pathways also regulate cognitive functions, we will discuss the evidence of ketamine's effect on cognitive function in MDD. Overall, the data accrued from pre-clinical research have implicated the mRNA translation pathways in treating mood symptoms of MDD. However, it is yet unclear whether the pro-cognitive potential of subanesthetic ketamine in rodents also engages these pathways and whether such an effect is consistently observed in the treatment-resistant MDD population.
Collapse
Affiliation(s)
- Vern Lewis
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Brandon Rodrigue
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Emily Arsenault
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Molly Zhang
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | | | | | - Mysa Myers
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Edna Matta-Camacho
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | | |
Collapse
|
15
|
Zhang R, Li D, Mao H, Wei X, Xu M, Zhang S, Jiang Y, Wang C, Xin Q, Chen X, Li G, Ji B, Yan M, Cai X, Dong B, Randeva HS, Liu C, Chen J. Disruption of 5-hydroxytryptamine 1A receptor and orexin receptor 1 heterodimer formation affects novel G protein-dependent signaling pathways and has antidepressant effects in vivo. Transl Psychiatry 2022; 12:122. [PMID: 35338110 PMCID: PMC8956632 DOI: 10.1038/s41398-022-01886-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/25/2022] [Accepted: 03/08/2022] [Indexed: 01/28/2023] Open
Abstract
G protein-coupled receptor (GPCR) heterodimers are new targets for the treatment of depression. Increasing evidence supports the importance of serotonergic and orexin-producing neurons in numerous physiological processes, possibly via a crucial interaction between 5-hydroxytryptamine 1A receptor (5-HT1AR) and orexin receptor 1 (OX1R). However, little is known about the function of 5-HT1AR/OX1R heterodimers. It is unclear how the transmembrane domains (TMs) of the dimer affect its function and whether its modulation mediates antidepressant-like effects. Here, we examined the mechanism of 5-HT1AR/OX1R dimerization and downstream G protein-dependent signaling. We found that 5-HT1AR and OX1R form constitutive heterodimers that induce novel G protein-dependent signaling, and that this heterodimerization does not affect recruitment of β-arrestins to the complex. In addition, we found that the structural interface of the active 5-HT1AR/OX1R dimer transforms from TM4/TM5 in the basal state to TM6 in the active conformation. We also used mutation analyses to identify key residues at the interface (5-HT1AR R1514.40, 5-HT1AR Y1985.41, and OX1R L2305.54). Injection of chronic unpredictable mild stress (CUMS) rats with TM4/TM5 peptides improved their depression-like emotional status and decreased the number of endogenous 5-HT1AR/OX1R heterodimers in the rat brain. These antidepressant effects may be mediated by upregulation of BDNF levels and enhanced phosphorylation and activation of CREB in the hippocampus and medial prefrontal cortex. This study provides evidence that 5-HT1AR/OX1R heterodimers are involved in the pathological process of depression. Peptides including TMs of the 5-HT1AR/OX1R heterodimer interface are candidates for the development of compounds with fast-acting antidepressant-like effects.
Collapse
Affiliation(s)
- Rumin Zhang
- grid.449428.70000 0004 1797 7280Neurobiology Institute, Jining Medical University, Jining, China
| | - Dandan Li
- grid.449428.70000 0004 1797 7280Neurobiology Institute, Jining Medical University, Jining, China
| | - Huiling Mao
- grid.449428.70000 0004 1797 7280Neurobiology Institute, Jining Medical University, Jining, China
| | - Xiaonan Wei
- grid.449428.70000 0004 1797 7280Neurobiology Institute, Jining Medical University, Jining, China
| | - MingDong Xu
- grid.449428.70000 0004 1797 7280Neurobiology Institute, Jining Medical University, Jining, China
| | - Shengnan Zhang
- grid.449428.70000 0004 1797 7280Neurobiology Institute, Jining Medical University, Jining, China
| | - Yunlu Jiang
- grid.449428.70000 0004 1797 7280Neurobiology Institute, Jining Medical University, Jining, China
| | - Chunmei Wang
- grid.449428.70000 0004 1797 7280Neurobiology Institute, Jining Medical University, Jining, China
| | - Qing Xin
- grid.449428.70000 0004 1797 7280Neurobiology Institute, Jining Medical University, Jining, China
| | - Xiaoyu Chen
- Department of Physiology, Shandong First Medical University, Taian, China
| | - Guorong Li
- grid.410585.d0000 0001 0495 1805School of Life Sciences, Shandong Normal University, Jinan, China
| | - Bingyuan Ji
- grid.449428.70000 0004 1797 7280Neurobiology Institute, Jining Medical University, Jining, China
| | - Maocai Yan
- grid.449428.70000 0004 1797 7280School of Pharmacy, Jining Medical University, Shandong, China
| | - Xin Cai
- grid.268079.20000 0004 1790 6079Department of Physiology, Weifang Medical University, Weifang, China
| | - Bo Dong
- grid.460018.b0000 0004 1769 9639Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Harpal S. Randeva
- grid.7372.10000 0000 8809 1613Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Chuanxin Liu
- grid.449428.70000 0004 1797 7280Neurobiology Institute, Jining Medical University, Jining, China
| | - Jing Chen
- Neurobiology Institute, Jining Medical University, Jining, China. .,Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK.
| |
Collapse
|
16
|
Cueto-Escobedo J, German-Ponciano LJ, Guillén-Ruiz G, Soria-Fregozo C, Herrera-Huerta EV. Zebrafish as a Useful Tool in the Research of Natural Products With Potential Anxiolytic Effects. Front Behav Neurosci 2022; 15:795285. [PMID: 35095438 PMCID: PMC8789748 DOI: 10.3389/fnbeh.2021.795285] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/16/2021] [Indexed: 11/13/2022] Open
Abstract
Zebrafish (Danio rerio) is a popular and valuable species used in many different biomedical research areas. The complex behavior that fish exhibit in response to different stimuli allows researchers to explore the biological and pharmacological basis of affective and mood disorders. In this sense, anxiety is commonly studied in preclinical research with animal models in rodents. During the last decade, those models have been successfully adapted to zebrafish. Stressful stimuli, such as novel environments, chemical substances, light conditions, and predator images, can trigger defensive behaviors considered indicators of an anxiety-like state. In the first stage, models were adapted and validated with different stressors and anxiolytic drugs with promising results and are now successfully used to generate scientific knowledge. In that sense, zebrafish allows several routes of administration and other methodological advantages to explore the anxiolytic effects of natural products in behavioral tests as novel tank, light-dark chamber, and black/white maze, among others. The present work will review the main findings on preclinical research using adult zebrafish to explore anxiolytics effects of natural products as plant secondary metabolites such as flavonoids, alkaloids and terpenes or standardized extracts of plants, among others. Scientific literature confirms the utility of zebrafish tests to explore anxiety-like states and anxiolytic-like effects of plant secondary metabolites, which represent a useful and ethical tool in the first stages of behavioral.
Collapse
Affiliation(s)
- Jonathan Cueto-Escobedo
- Departamento de Investigación Clínica y Traslacional, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | | | - Gabriel Guillén-Ruiz
- Investigador por México, Consejo Nacional de Ciencia y Tecnología (CONACyT) – Instituto de Neuroetología, Universidad Veracruzana, Xalapa, Mexico
| | - Cesar Soria-Fregozo
- Laboratorio Ciencias Biomédicas/Área Histología y Psicobiología, Departamento de Ciencias de la Tierra y de la Vida, Centro Universitario de Los Lagos, Universidad de Guadalajara, Lagos de Moreno, Mexico
| | | |
Collapse
|
17
|
Hatakama H, Asaoka N, Nagayasu K, Shirakawa H, Kaneko S. A selective serotonin reuptake inhibitor ameliorates obsessive-compulsive disorder-like perseverative behavior by attenuating 5-HT 2C receptor signaling in the orbitofrontal cortex. Neuropharmacology 2021; 206:108926. [PMID: 34921828 DOI: 10.1016/j.neuropharm.2021.108926] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/20/2021] [Accepted: 12/13/2021] [Indexed: 10/19/2022]
Abstract
Perseveration is a characteristic of patients with obsessive-compulsive disorder (OCD). Clinically, neuronal activity in the lateral orbitofrontal cortex (OFC) is increased in OCD patients. Successful treatment with selective serotonin reuptake inhibitors (SSRIs) reduces activity in the lateral OFC of OCD patients, but the precise mechanisms underlying this effect are unclear. Previously, we reported that repeated injection of the dopamine D2 receptor agonist quinpirole (QNP) resulted in OCD-like deficits, including perseveration in a reversal learning task. QNP-treated mice showed hyperactivity in lateral OFC pyramidal neurons. The present study demonstrated that 4-week administration of an SSRI increased the rate of correct choice in a reversal learning task. Using the electrophysiological approach, we revealed that an SSRI decreased the activity of lateral OFC pyramidal neurons in QNP-treated mice by potentiating inhibitory inputs. The 4-week administration of an SSRI inhibited the potentiation of neuronal activity induced by a 5-HT2C receptor agonist. Additionally, both 4-week administration of SSRI and acute application of 5-HT2C receptor antagonist prevented the QNP-induced potentiation of inhibitory inputs to fast-spiking interneurons in the lateral OFC. Administration of a 5-HT2C receptor antagonist to mice for 4 days increased the rate of correct choice in a reversal learning task. Collectively, these results indicate that chronic SSRI ameliorated perseverative behavior in QNP-treated mice by modulating inhibitory inputs in the lateral OFC. Short-term 5-HT2C receptor blockade also ameliorated QNP-induced behavioral and neurological abnormalities by, at least in part, a common mechanism with chronic SSRI.
Collapse
Affiliation(s)
- Hikari Hatakama
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Nozomi Asaoka
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan; Department of Pharmacology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
18
|
Astroglial Serotonin Receptors as the Central Target of Classic Antidepressants. ADVANCES IN NEUROBIOLOGY 2021; 26:317-347. [PMID: 34888840 DOI: 10.1007/978-3-030-77375-5_13] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Major depressive disorder (MDD) presents multiple clinical phenotypes and has complex underlying pathological mechanisms. Existing theories cannot completely explain the pathophysiological mechanism(s) of MDD, while the pharmacology of current antidepressants is far from being fully understood. Astrocytes, the homeostatic and defensive cells of the central nervous system, contribute to shaping behaviors, and regulating mood and emotions. A detailed introduction on the role of astrocytes in depressive disorders is thus required, to which this chapter is dedicated. We also focus on the interactions between classic antidepressants and serotonin receptors, overview the role of astrocytes in the pharmacological mechanisms of various antidepressants, and present astrocytes as targets for the treatment of bipolar disorder. We provide a foundation of knowledge on the role of astrocytes in depressive disorders and astroglial 5-HT2B receptors as targets for selective serotonin reuptake inhibitors in vivo and in vitro.
Collapse
|
19
|
Chen T, Wang R, Duan Z, Yuan X, Ding Y, Feng Z, Bu F, Liu L, Wang Q, Zhou J, Zhu L, Ni Q, Shi G, Chen Y. Akkermansia muciniphila Protects Against Psychological Disorder-Induced Gut Microbiota-Mediated Colonic Mucosal Barrier Damage and Aggravation of Colitis. Front Cell Infect Microbiol 2021; 11:723856. [PMID: 34722332 PMCID: PMC8551916 DOI: 10.3389/fcimb.2021.723856] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 09/20/2021] [Indexed: 12/22/2022] Open
Abstract
Psychological disorders are associated with increased risk of severe inflammatory bowel disease (IBD) by causing gut microbiota dysbiosis and colonic mucosal barrier damage. However, the interaction between chronic restraint stress (CRS), gut microbiota composition, and colonic mucus remains unclear. We demonstrated that mice under CRS conditions exhibited alterations in microbiota composition, disruption of colonic mucus, and aggravation of colitis. In addition, the abundance of Akkermansia muciniphila was significantly decreased in mice under CRS and UC patients with depression, and positively associated with the expression of MUC2. After antibiotic treatment, the recipient mice colonized with CRS microbiota showed barrier defects and severe colitis. Administration of Akkermansia muciniphila was found to restore colonic mucus and modify the gut microbiota. We confirm that CRS-mediated gut microbiota dysbiosis results in colonic mucosal barrier damage and aggravation of colitis. Our results suggest that A. muciniphila is expected to be a potential probiotic to protect and treat colonic mucus that is involved in IBD with psychological disorders.
Collapse
Affiliation(s)
- Tuo Chen
- Department of General Surgery, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Rong Wang
- Department of Colorectal Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhenglan Duan
- Department of Colorectal Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaomin Yuan
- Department of Colorectal Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Ding
- Department of Colorectal Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zeyu Feng
- Department of Colorectal Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Fan Bu
- Department of Colorectal Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Liu
- Glycomics and Glycan Bioengineering Research Center, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Qiong Wang
- Basic Pharmacology Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinyong Zhou
- Central Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lei Zhu
- Collaborative Innovation Center for Cancer Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qing Ni
- Department of General Surgery, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Guoping Shi
- Collaborative Innovation Center for Cancer Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yugen Chen
- Department of Colorectal Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
20
|
Biney RP, Benneh CK, Adongo DW, Ameyaw EO, Woode E. Evidence of an antidepressant-like effect of xylopic acid mediated by serotonergic mechanisms. Psychopharmacology (Berl) 2021; 238:2105-2120. [PMID: 33837810 DOI: 10.1007/s00213-021-05835-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/22/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Depression causes significant debilitating symptoms and economic burden. Current management is challenged by slow onset of action and modest efficacies of antidepressants; thus, the search for newer antidepressants remains relevant. We evaluated the antidepressant effects of a kaurene diterpene, xylopic acid (XA), in zebrafish and mouse models. METHODS The chronic unpredictable stress (CUS) protocol in zebrafish and the tail suspension test (TST), forced swim test (FST), lipopolysaccharide-induced depression-like behaviour test (LID) and repeated open space swimming test (OSST) in mice were used. We further examined the impact of depleting monoamines on XA's antidepressant effects. The contribution of glutamatergic and nitrergic pathways on the antidepressant effect of XA in mice and XA's effects on 5-HT receptors and monoamine oxidase (MAO) enzymes were also evaluated. Finally, XA's influence on neuroprotection was evaluated by measuring BDNF and oxidative stress enzymes in whole brain. XA doses (1-10 μM) in zebrafish and (10, 30, 100 mg kg-1) in mice exerted potent antidepressant-like potential in FST, TST, LID and showed fast-onset antidepressant-like property in the OSST. RESULTS The antidepressant-like properties in mice were reversed by blocking synthesis/release of serotonin but not noradrenaline using p-chlorophenylalanine and α-methyl-p-tyrosine, respectively. This antidepressant-like effect was potentiated by D-cycloserine and Nω-Nitro-L-arginine methyl ester (L-NAME) but not by D-serine and L-arginine. XA also evoked partial agonist-like effects on 5-hydroxytrptamine receptors on the rat fundus but it did not have MAO inhibition effect. It also increased BDNF, glutathione and antioxidant enzymes. CONCLUSION Therefore, xylopic acid possesses antidepressant-like effects largely mediated by serotonergic and neuroprotective mechanisms.
Collapse
Affiliation(s)
- Robert Peter Biney
- Department of Pharmacology, School of Medical Sciences, University of Cape Coast, Cape Coast, Ghana.
- School of Pharmacy and Pharmaceutical Sciences, University of Cape Coast, Cape Coast, Ghana.
| | - Charles Kwaku Benneh
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| | - Donatus Wewura Adongo
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
| | - Elvis Ofori Ameyaw
- School of Pharmacy and Pharmaceutical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Eric Woode
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Health and Allied Sciences, Ho, Ghana
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
21
|
Early response to trazodone once-a-day in major depressive disorder: review of the clinical data and putative mechanism for faster onset of action. CNS Spectr 2021; 26:232-242. [PMID: 33731232 DOI: 10.1017/s1092852921000304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Most antidepressants have a delayed onset of action and must be administered for several weeks to generate therapeutic effects. Trazodone is a serotonin antagonist and reuptake inhibitor approved for the treatment of major depressive disorder. The once-a-day (OAD) formulation of trazodone has an improved tolerability profile compared to its conventional formulations. In this study, we systematically reviewed the evidence available for the antidepressant efficacy and early improvement in depressive symptoms with trazodone OAD treatment. METHOD We conducted a PubMed database search for randomized controlled trials published from 2005 to 2020. RESULTS Two studies, a placebo-controlled and an active-comparator (venlafaxine extended-release or XR) study were found. Both the studies demonstrated that trazodone exhibits antidepressant activity at a starting dose of 150 mg/day and results in statistically significant greater reduction in Hamilton Depression Rating Scale (HAM-D17) scores within 1 week of starting treatment compared to placebo or venlafaxine XR (P < .05). Trazodone also resulted in significant early improvement in the HAM-D17 sleep disturbance factor compared to placebo or venlafaxine XR at day 7 (P < .05). This clinical effect is supported by in vitro proprietary data for the affinity of trazodone for different target receptors. Activity at these receptors may underlie trazodone's fast antidepressant action. CONCLUSIONS Trazodone, if properly dosed, can be an effective antidepressant with early onset of action and good tolerability. Future studies designed to specifically evaluate onset and timing of improvement of depressive symptoms remain necessary to confirm and extend these results.
Collapse
|
22
|
EEG and Sleep Effects of Tramadol Suggest Potential Antidepressant Effects with Different Mechanisms of Action. Pharmaceuticals (Basel) 2021; 14:ph14050431. [PMID: 34064349 PMCID: PMC8147808 DOI: 10.3390/ph14050431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 01/18/2023] Open
Abstract
Tramadol is a widely used, centrally acting, opioid analgesic compound, with additional inhibitory effects on the synaptic reuptake of serotonin and noradrenaline, as well as on the 5-HT2 and NMDA receptors. Preclinical and clinical evidence also suggests its therapeutic potential in the treatment of depression and anxiety. The effects of most widely used antidepressants on sleep and quantitative electroencephalogram (qEEG) are well characterized; however, such studies of tramadol are scarce. Our aim was to characterize the effects of tramadol on sleep architecture and qEEG in different sleep–wake stages. EEG-equipped Wistar rats were treated with tramadol (0, 5, 15 and 45 mg/kg) at the beginning of the passive phase, and EEG, electromyogram and motor activity were recorded. Tramadol dose-dependently reduced the time spent in rapid eye movement (REM) sleep and increased the REM onset latency. Lower doses of tramadol had wake-promoting effects in the first hours, while 45 mg/kg of tramadol promoted sleep first, but induced wakefulness thereafter. During non-REM sleep, tramadol (15 and 45 mg/kg) increased delta and decreased alpha power, while all doses increased gamma power. In conclusion, the sleep-related and qEEG effects of tramadol suggest antidepressant-like properties, including specific beneficial effects in selected patient groups, and raise the possibility of a faster acting antidepressant action.
Collapse
|
23
|
Gao N, Zheng W, Murezati T, Gu W, Li X, Jin Z. GW117: A novel serotonin (5-HT 2C ) receptor antagonist and melatonin (MT 1 /MT 2 ) receptor agonist with potential antidepressant-like activity in rodents. CNS Neurosci Ther 2021; 27:702-713. [PMID: 33650297 PMCID: PMC8111499 DOI: 10.1111/cns.13630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/28/2022] Open
Abstract
Aims To evaluate the antidepressant‐like effect of compound GW117 in rodents using in vitro binding and uptake assays as well in vivo behavioral tests. Methods We investigated the target profile of GW117 using [35S]‐GTPγS and [3H]PIP binding. Using the forced swimming test and chronic unpredictable stress in rats, tail suspension test in mice and rats, and learned helplessness model in mice, we further revealed the antidepressant‐like and anxiolytic‐like effects of GW117. Results The current study suggests that GW117 displays serotonin 2C (5‐HT2C) receptor antagonist and melatonin type 1 and 2 (MT1/MT2) receptor agonist properties, as well as evident antidepressant and anxiolytic effects. Conclusion These data suggest that GW117 is probably a potent antidepressant.
Collapse
Affiliation(s)
- Nana Gao
- Department of Gastrointestinal Surgery and Clinical Nutrition, Beijing Shijitan Hospital,Captial Medical University, Beijing, China
| | - Wei Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Tiliwaerde Murezati
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wei Gu
- Beijing Guangwei Pharmaceutical Technology Co., Ltd, Beijing, China
| | - Xiaorong Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zengliang Jin
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
De Gregorio D, Popic J, Enns JP, Inserra A, Skalecka A, Markopoulos A, Posa L, Lopez-Canul M, He Q, Lafferty CK, Britt JP, Comai S, Aguilar-Valles A, Sonenberg N, Gobbi G. Lysergic acid diethylamide (LSD) promotes social behavior through mTORC1 in the excitatory neurotransmission. Proc Natl Acad Sci U S A 2021; 118:e2020705118. [PMID: 33495318 PMCID: PMC7865169 DOI: 10.1073/pnas.2020705118] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Clinical studies have reported that the psychedelic lysergic acid diethylamide (LSD) enhances empathy and social behavior (SB) in humans, but its mechanism of action remains elusive. Using a multidisciplinary approach including in vivo electrophysiology, optogenetics, behavioral paradigms, and molecular biology, the effects of LSD on SB and glutamatergic neurotransmission in the medial prefrontal cortex (mPFC) were studied in male mice. Acute LSD (30 μg/kg) injection failed to increase SB. However, repeated LSD (30 μg/kg, once a day, for 7 days) administration promotes SB, without eliciting antidepressant/anxiolytic-like effects. Optogenetic inhibition of mPFC excitatory neurons dramatically inhibits social interaction and nullifies the prosocial effect of LSD. LSD potentiates the α-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) and 5-HT2A, but not N-methyl-D-aspartate (NMDA) and 5-HT1A, synaptic responses in the mPFC and increases the phosphorylation of the serine-threonine protein kinases Akt and mTOR. In conditional knockout mice lacking Raptor (one of the structural components of the mTORC1 complex) in excitatory glutamatergic neurons (Raptorf/f:Camk2alpha-Cre), the prosocial effects of LSD and the potentiation of 5-HT2A/AMPA synaptic responses were nullified, demonstrating that LSD requires the integrity of mTORC1 in excitatory neurons to promote SB. Conversely, in knockout mice lacking Raptor in GABAergic neurons of the mPFC (Raptorf/f:Gad2-Cre), LSD promotes SB. These results indicate that LSD selectively enhances SB by potentiating mPFC excitatory transmission through 5-HT2A/AMPA receptors and mTOR signaling. The activation of 5-HT2A/AMPA/mTORC1 in the mPFC by psychedelic drugs should be explored for the treatment of mental diseases with SB impairments such as autism spectrum disorder and social anxiety disorder.
Collapse
Affiliation(s)
- Danilo De Gregorio
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada, H3A 1A1;
- Department of Biochemistry, McGill University, Montreal, QC, Canada, H3A 1A3
| | - Jelena Popic
- Department of Biochemistry, McGill University, Montreal, QC, Canada, H3A 1A3
| | - Justine P Enns
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada, H3A 1A1
| | - Antonio Inserra
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada, H3A 1A1
| | - Agnieszka Skalecka
- Department of Biochemistry, McGill University, Montreal, QC, Canada, H3A 1A3
| | - Athanasios Markopoulos
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada, H3A 1A1
| | - Luca Posa
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada, H3A 1A1
| | - Martha Lopez-Canul
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada, H3A 1A1
| | - Qianzi He
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada, H3A 1A1
| | | | - Jonathan P Britt
- Department of Psychology, McGill University, Montreal, QC, Canada, H3A 1B1
| | - Stefano Comai
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada, H3A 1A1
- Division of Neuroscience, Vita Salute San Raffaele University, 20132 Milan, Italy
| | | | - Nahum Sonenberg
- Department of Biochemistry, McGill University, Montreal, QC, Canada, H3A 1A3;
| | - Gabriella Gobbi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, QC, Canada, H3A 1A1;
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada, K1S 5B6
| |
Collapse
|
25
|
Jefferson SJ, Feng M, Chon UR, Guo Y, Kim Y, Luscher B. Disinhibition of somatostatin interneurons confers resilience to stress in male but not female mice. Neurobiol Stress 2020; 13:100238. [PMID: 33344694 PMCID: PMC7739040 DOI: 10.1016/j.ynstr.2020.100238] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/22/2020] [Accepted: 07/01/2020] [Indexed: 12/16/2022] Open
Abstract
Chronic stress represents a vulnerability factor for anxiety and depressive disorders and has been widely used to model aspects of these disorders in rodents. Disinhibition of somatostatin (SST)-positive GABAergic interneurons in mice by deletion of γ2 GABAA receptors selectively from these cells (SSTCre:γ2f/f mice) has been shown to result in behavioral and biochemical changes that mimic the responses to antidepressant doses of ketamine. Here we explored the extent to which SSTCre:γ2f/f mice exhibit resilience to unpredictable chronic mild stress (UCMS). We found that male SSTCre:γ2f/f mice are resilient to UCMS-induced (i) reductions in weight gain, (ii) reductions in SST-immuno-positive cells in medial prefrontal cortex (mPFC), (iii) increases in phosphorylation of eukaryotic elongation factor 2 (eEF2) in mPFC, and (iv) increased anxiety in a novelty suppressed feeding test. Female SSTCre:γ2f/f mice were resilient to UCMS-induced reductions in SST-immuno-positive cells indistinguishably from males. However, in contrast to males, they showed no UCMS effects on weight gain independent of genotype. Moreover, in mPFC of female γ2f/f control mice, UCMS resulted in paradoxically reduced p-EF2 levels without stress effects in the SSTCre:γ2f/f mutants. Lastly, female SSTCre:γ2f/f mice showed increased rather than reduced UCMS induced anxiety compared to γ2f/f controls. Thus, disinhibition of SST interneurons results in behavioral resilience to UCMS selectively in male mice, along with cellular resilience of SST neurons to UCMS independent of sex. Thus, mechanisms underlying vulnerability and resilience to stress are sex specific and map to mPFC rather than hippocampus but appear unrelated to changes in expression of SST as a marker of corresponding interneurons.
Collapse
Affiliation(s)
- Sarah J. Jefferson
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA
- Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Mengyang Feng
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA
- Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - URee Chon
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, 17033, USA
| | - Yao Guo
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA
- Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Yongsoo Kim
- Department of Neural and Behavioral Sciences, College of Medicine, Pennsylvania State University, Hershey, PA, 17033, USA
| | - Bernhard Luscher
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA, 16802, USA
- Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
26
|
Rapid acting antidepressants in the mTOR pathway: Current evidence. Brain Res Bull 2020; 163:170-177. [DOI: 10.1016/j.brainresbull.2020.07.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/16/2020] [Accepted: 07/24/2020] [Indexed: 02/08/2023]
|
27
|
Cueto-Escobedo J, Andrade-Soto J, Lima-Maximino M, Maximino C, Hernández-López F, Rodríguez-Landa JF. Involvement of GABAergic system in the antidepressant-like effects of chrysin (5,7-dihydroxyflavone) in ovariectomized rats in the forced swim test: comparison with neurosteroids. Behav Brain Res 2020; 386:112590. [DOI: 10.1016/j.bbr.2020.112590] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 01/28/2020] [Accepted: 02/21/2020] [Indexed: 10/24/2022]
|
28
|
Wu R, Xiao D, Shan X, Dong Y, Tao WW. Rapid and Prolonged Antidepressant-like Effect of Crocin Is Associated with GHSR-Mediated Hippocampal Plasticity-related Proteins in Mice Exposed to Prenatal Stress. ACS Chem Neurosci 2020; 11:1159-1170. [PMID: 32203651 DOI: 10.1021/acschemneuro.0c00022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Prenatal stress (PNS) has a prolonged and adverse effect on offspring, leading to a significantly increased vulnerability to developing depression in their later life. Traditional therapies have delayed onset and limited efficacy; thus, it remains an urgent need to find novel medications with fast-onset and high-efficacy potentials. Crocin, with its structure clearly examined, has shown antidepressant-like effects. However, few studies extensively investigated its effect especially in mice exposed to PNS. Using an established PNS model, we tested whether crocin could have a rapid and persistent antidepressant-like effect in PNS mice. Growth hormone secretagogue receptor (GHSR) and phosphoinositide 3-kinase (PI3K) inhibitors were used to test their effects in antidepressant-like effect of crocin. Hippocampal GHSR-PI3K signaling was examined both in PNS mice treated with a single dose of crocin and in combination of GHSR inhibitor. PNS mice showed depression-like behaviors at juvenile and adulthood, and crocin induced an instant and persistent antidepressant-like response in PNS mice in a dose-dependent manner. Moreover, crocin increased the expression of hippocampal synaptic plasticity-associated proteins through the restoration of GHSR-PI3K signaling. Inhibitions of both GHSR and PI3K abolished the effect of crocin in alleviating depressive-like behaviors. More importantly, GHSR inhibitor JMV2959 blocked the enhanced expression of hippocampal plasticity-related proteins induced by crocin. The present study demonstrated that crocin induced a fast-onset and prolonged antidepressant effect in PNS mice and suggested that GHSR-PI3K signaling may play a key role in crocin's effect at least partially by a restoration of hippocampal synaptic plasticity-associated proteins.
Collapse
Affiliation(s)
- Ruyan Wu
- School of Medicine, Yangzhou University, Yangzhou 225000, China
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo 14203, New York, United States
| | - Dong Xiao
- Key Laboratory of Integrative Biomedicine for Brain Diseases, School of Basic Biomedical Science, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xin Shan
- Key Laboratory of Integrative Biomedicine for Brain Diseases, School of Basic Biomedical Science, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Dong
- Key Laboratory of Integrative Biomedicine for Brain Diseases, School of Basic Biomedical Science, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei-Wei Tao
- Key Laboratory of Integrative Biomedicine for Brain Diseases, School of Basic Biomedical Science, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
29
|
Xiao Y, Luo H, Yang WZ, Zeng Y, Shen Y, Ni X, Shi Z, Zhong J, Liang Z, Fu X, Tu H, Sun W, Shen WL, Hu J, Yang J. A Brain Signaling Framework for Stress-Induced Depression and Ketamine Treatment Elucidated by Phosphoproteomics. Front Cell Neurosci 2020; 14:48. [PMID: 32317933 PMCID: PMC7156020 DOI: 10.3389/fncel.2020.00048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/20/2020] [Indexed: 12/25/2022] Open
Abstract
Depression is a common affective disorder characterized by significant and persistent low mood. Ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist, is reported to have a rapid and durable antidepressant effect, but the mechanisms are unclear. Protein phosphorylation is a post-translational modification that plays a crucial role in cell signaling. Thus, we present a phosphoproteomics approach to investigate the mechanisms underlying stress-induced depression and the rapid antidepressant effect of ketamine in mice. We analyzed the phosphoprotein changes induced by chronic unpredictable mild stress (CUMS) and ketamine treatment in two known mood control centers, the medial prefrontal cortex (mPFC) and the nucleus accumbens (NAc). We initially obtained >8,000 phosphorylation sites. Quantitation revealed 3,988 sites from the mPFC and 3,196 sites from the NAc. Further analysis revealed that changes in synaptic transmission-related signaling are a common feature. Notably, CUMS-induced changes were reversed by ketamine treatment, as shown by the analysis of commonly altered sites. Ketamine also induced specific changes, such as alterations in synapse organization, synaptic transmission, and enzyme binding. Collectively, our findings establish a signaling framework for stress-induced depression and the rapid antidepressant effect of ketamine.
Collapse
Affiliation(s)
- Yan Xiao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Huoqing Luo
- School of Life Science and Technology, Shanghaitech University, Shanghai, China.,State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Institute of Neuroscience, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wen Z Yang
- Shanghai Institute for Advanced Immunochemical Studies & School of Life Science and Technology, Shanghaitech University, Shanghai, China.,CAS Key Laboratory of Synthetic Chemistry of Natural Substances, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Yeting Zeng
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yinbo Shen
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xinyan Ni
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Zhaomei Shi
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Jun Zhong
- Delta Omics Inc., Baltimore, MD, United States
| | - Ziqi Liang
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Xiaoyu Fu
- School of Life Science and Technology, Shanghaitech University, Shanghai, China.,State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Institute of Neuroscience, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hongqing Tu
- School of Life Science and Technology, Shanghaitech University, Shanghai, China.,State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Institute of Neuroscience, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenzhi Sun
- Chinese Institute For Brain Research, Beijing, China
| | - Wei L Shen
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Ji Hu
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Jiajun Yang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
30
|
Shang C, Yao RM, Guo Y, Ding ZC, Sun LJ, Ran YH, Xue R, Wang HS, Zhang JM, Zhang YZ, Zhang LM, Li YF. Translocator protein-mediated fast-onset antidepressant-like and memory-enhancing effects in chronically stressed mice. J Psychopharmacol 2020; 34:441-451. [PMID: 31913078 DOI: 10.1177/0269881119896304] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Fast-acting and cognitive-enhancing antidepressants are desperately needed. Activation of translocator protein (18 kDa, TSPO) is a novel strategy for developing potential antidepressants, but there are no data available on the onset time of TSPO ligands. This study aimed to investigate the fast-onset antidepressant actions of AC-5216, a selective TSPO ligand, in TSPO knock-out (KO) mice. METHODS TSPO wild-type (WT) and KO mice were subjected to a six-week chronic unpredicted stress (CUS) paradigm. Then, the mice were treated with AC-5216 and tested with depressive and cognitive behaviours. RESULTS A single dose of AC-5216 (0.3 mg/kg) exerted anxiolytic- and antidepressant-like actions in TSPO WT mice. Moreover, in chronically stressed WT mice, two to four days of AC-5216 treatment (0.3 mg/kg, once per day) produced fast-onset antidepressant-like effects in the novelty-suppressed feeding and sucrose preference tests, as well as memory-enhancing effects in the novel object recognition test. In addition, a rapid (with five days of treatment) restoration of serum corticosterone levels and prefrontal cortex (PFC) allopregnanolone levels was found. Further studies showed that in these stress-exposed WT mice, AC-5216 significantly increased the levels of mTOR signalling-related proteins (mBDNF, p-mTOR, PSD-95, synapsin-1, GluR1), as well as the total dendritic length and branching points of pyramidal neurons in the PFC. CONCLUSIONS These results suggest that TSPO mediates the fast-onset antidepressant-like and memory-enhancing effects of AC-5216, possibly through the rapid activation of mTOR signalling and restoration of dendritic complexity in the PFC.
Collapse
Affiliation(s)
- Chao Shang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Ru-Meng Yao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Ying Guo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Zhen-Chun Ding
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Li-Jun Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Yu-Hua Ran
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Rui Xue
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Huai-Shan Wang
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, PR China
| | - Jian-Min Zhang
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, State Key Laboratory of Medical Molecular Biology, Beijing, PR China
| | - You-Zhi Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Li-Ming Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| | - Yun-Feng Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, PR China
| |
Collapse
|
31
|
Ma X, Zhu Z, Guo S, Duan J. The effect of deoxyschizandrin on chronic unpredictable mild stress-induced depression. Biotechnol Appl Biochem 2020; 68:52-59. [PMID: 31985079 DOI: 10.1002/bab.1893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 01/23/2020] [Indexed: 01/17/2023]
Abstract
The purpose of the present study was to evaluate the antidepressant effect of deoxyschizandrin (DEO) in chronic unpredictable mild stress (CUMS)-induced mice. The mice were subjected to CUMS paradigm for 8 weeks. From the sixth week, the mice were intragastrically treated with DEO once daily for continuous 3 weeks. The behavior tests including sucrose preference test (SPT), forced swimming test (FST), tail suspension test (TST), and open field test were conducted. Additionally, the expressions of TLR4, MyD88, TRAF6, p-NF-κBp65, NLRP3, cleaved caspase-1, cleaved IL-1β, GluR, and PSD95 in hippocampus were detected by western blot. The concentrations of IL-6 and TNF-α in hippocampus were determined by enzyme linked immune sorbent assay (ELISA). The dendritic spine density was observed by Golgi-Cox staining. As a result, the treatment with DEO relieved anhedonia in SPT, and reduced immobile duration in FST and TST. DEO treatment effectively attenuated the CUMS-caused alterations of TLR4, MyD88, TRAF6, p-NF-κBp65, NLRP3, cleaved caspase-1, cleaved IL-1β, GluR, and PSD95. Furthermore, DEO could reduce the hippocampal inflammatory cytokine content and increase the density of dendritic spine. In conclusion, the present work indicated that DEO exhibited antidepressant effect on CUMS-induced depressive mice, which was possible due to the TLR4/NF-κB/NLRP3 pathway and the amelioration of dendritic spine density through GluR/PSD95 cascade.
Collapse
Affiliation(s)
- Xinfei Ma
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhenhua Zhu
- Institute of Mental Health, Suzhou Psychiatric Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Sheng Guo
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
32
|
Kim J, Cha E, Park WK, Lee HY, Lim SM, Kim HJ, Pae AN. Evaluation of anti-depressant effects of phthalazinone-based triple-acting small molecules against 5-HT2A, 5-HT2C, and the serotonin transporter. Bioorg Med Chem Lett 2020; 30:126882. [DOI: 10.1016/j.bmcl.2019.126882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/27/2019] [Accepted: 12/01/2019] [Indexed: 12/15/2022]
|
33
|
Papp N, Koncz S, Kostyalik D, Kitka T, Petschner P, Vas S, Bagdy G. Acute 5-HT 2C Receptor Antagonist SB-242084 Treatment Affects EEG Gamma Band Activity Similarly to Chronic Escitalopram. Front Pharmacol 2020; 10:1636. [PMID: 32063851 PMCID: PMC7000428 DOI: 10.3389/fphar.2019.01636] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022] Open
Abstract
Serotonin 2C receptors (5-HT2CRs) are implicated in the pathomechanism and treatment of anxiety and depression. Recently, as a new biomarker of depression, alterations in the gamma power of the electroencephalogram (EEG) have been suggested. Chronic treatment with the selective serotonin reuptake inhibitor (SSRI) antidepressant escitalopram has been shown to cause sleep-wake stage-dependent alterations in gamma power. However, despite the antidepressant potency of 5-HT2CR-antagonists, there is no data available regarding the effects of selective 5-HT2CR-antagonists on gamma activity. Therefore, we investigate the acute effect of the 5-HT2CR-antagonist SB-242084 on gamma power in different vigilance stages when given in monotherapy, or in combination with chronic escitalopram treatment. We administered SB-242084 (1 mg/kg, intraperitoneally) or vehicle to EEG-equipped rats after a 21-day-long pretreatment with escitalopram (10 mg/kg/day, via osmotic minipumps) or vehicle. Frontoparietal EEG, electromyogram, and motor activity were recorded during the first 3 h of passive phase, after the administration of SB-242084. Quantitative EEG analysis revealed that acute SB-242084 increased gamma power (30-60 Hz) in light and deep slow-wave sleep, and passive wakefulness. However, in active wakefulness, rapid eye movement sleep, and intermediate stage, no change was observed in gamma power. The profile of the effect of SB-242084 on gamma power was similar to that produced by chronic escitalopram. Moreover, SB-242084 did not alter chronic escitalopram-induced effects on gamma. In conclusion, the similarity in the effect of the 5-HT2CR-antagonist and chronic SSRI on gamma power provides further evidence for the therapeutic potential of 5-HT2CR-antagonists in the treatment of depression and/or anxiety.
Collapse
Affiliation(s)
- Noémi Papp
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
| | - Szabolcs Koncz
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
| | - Diána Kostyalik
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
| | - Tamás Kitka
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
| | - Péter Petschner
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary.,MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Budapest, Hungary
| | - Szilvia Vas
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary.,MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Budapest, Hungary.,Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - György Bagdy
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary.,MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Budapest, Hungary.,NAP-2-SE New Antidepressant Target Research Group, Budapest, Hungary
| |
Collapse
|
34
|
5-HT2C receptor blockade reverses SSRI-associated basal ganglia dysfunction and potentiates therapeutic efficacy. Mol Psychiatry 2020; 25:3304-3321. [PMID: 30120415 PMCID: PMC6378140 DOI: 10.1038/s41380-018-0227-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 07/13/2018] [Accepted: 07/24/2018] [Indexed: 11/09/2022]
Abstract
Serotonin (5-HT) selective reuptake inhibitors (SSRIs) are widely used in the treatment of depression and anxiety disorders, but responsiveness is uncertain and side effects often lead to discontinuation. Side effect profiles suggest that SSRIs reduce dopaminergic (DAergic) activity, but specific mechanistic insight is missing. Here we show in mice that SSRIs impair motor function by acting on 5-HT2C receptors in the substantia nigra pars reticulata (SNr), which in turn inhibits nigra pars compacta (SNc) DAergic neurons. SSRI-induced motor deficits can be reversed by systemic or SNr-localized 5-HT2C receptor antagonism. SSRIs induce SNr hyperactivity and SNc hypoactivity that can also be reversed by systemic 5-HT2C receptor antagonism. Optogenetic inhibition of SNc DAergic neurons mimics the motor deficits due to chronic SSRI treatment, whereas local SNr 5-HT2C receptor antagonism or optogenetic activation of SNc DAergic neurons reverse SSRI-induced motor deficits. Lastly, we find that 5-HT2C receptor antagonism potentiates the antidepressant and anxiolytic effects of SSRIs. Together our findings demonstrate opposing roles for 5-HT2C receptors in the effects of SSRIs on motor function and affective behavior, highlighting the potential benefits of 5-HT2C receptor antagonists for both reduction of motor side effects of SSRIs and augmentation of therapeutic antidepressant and anxiolytic effects.
Collapse
|
35
|
Kraus C, Wasserman D, Henter ID, Acevedo-Diaz E, Kadriu B, Zarate CA. The influence of ketamine on drug discovery in depression. Drug Discov Today 2019; 24:2033-2043. [PMID: 31382015 PMCID: PMC6842425 DOI: 10.1016/j.drudis.2019.07.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/24/2019] [Accepted: 07/26/2019] [Indexed: 12/20/2022]
Abstract
Recent research demonstrating that the glutamatergic modulator ketamine has rapid, robust, and sustained antidepressant effects has been a turning point in drug discovery for depression. The recent FDA approval of esketamine for adults with treatment-resistant major depressive disorder (MDD) has further underscored the relevance of this agent in spurring investigation into novel and mechanistically distinct agents for use in depression. Over the past two decades, ketamine research has ushered in a new wave of studies seeking to not only identify its mechanism of action but also to examine the antidepressant potential of novel or repurposed agents. This article reviews the approaches that have proven particularly fruitful for the field of neuropsychiatry.
Collapse
Affiliation(s)
- Christoph Kraus
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Daniel Wasserman
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Ioline D Henter
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Elia Acevedo-Diaz
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| | - Bashkim Kadriu
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, USA.
| | - Carlos A Zarate
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, USA
| |
Collapse
|
36
|
Xiao D, Liu L, Li Y, Ruan J, Wang H. Licorisoflavan A Exerts Antidepressant-Like Effect in Mice: Involvement of BDNF-TrkB Pathway and AMPA Receptors. Neurochem Res 2019; 44:2044-2056. [DOI: 10.1007/s11064-019-02840-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/23/2019] [Accepted: 06/30/2019] [Indexed: 12/15/2022]
|
37
|
Modulation of Monoaminergic Systems by Antidepressants in the Frontal Cortex of Rats After Chronic Mild Stress Exposure. Mol Neurobiol 2019; 56:7522-7533. [DOI: 10.1007/s12035-019-1619-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/23/2019] [Indexed: 12/13/2022]
|
38
|
Crowley NA, Magee SN, Feng M, Jefferson SJ, Morris CJ, Dao NC, Brockway DF, Luscher B. Ketamine normalizes binge drinking-induced defects in glutamatergic synaptic transmission and ethanol drinking behavior in female but not male mice. Neuropharmacology 2019; 149:35-44. [PMID: 30731135 DOI: 10.1016/j.neuropharm.2019.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/30/2019] [Accepted: 02/02/2019] [Indexed: 01/06/2023]
Abstract
Ketamine is a fast acting experimental antidepressant with significant therapeutic potential for emotional disorders such as major depressive disorder and alcohol use disorders. Of particular interest is binge alcohol use, which during intermittent withdrawal from drinking involves depressive-like symptoms reminiscent of major depressive disorder. Binge drinking has been successfully modeled in mice with the Drinking in the Dark (DID) paradigm, which involves daily access to 20% ethanol, for a limited duration and selectively during the dark phase of the circadian light cycle. Here we demonstrate that DID exposure reduces the cell surface expression of NMDA- and AMPA-type glutamate receptors in the prelimbic cortex (PLC) of female but not male mice, along with reduced activity of the mammalian target of rapamycin (mTOR) signaling pathway. Pretreatment with an acute subanesthetic dose of ketamine suppresses binge-like ethanol consumption in female but not male mice. Lastly, DID-exposure reduces spontaneous glutamatergic synaptic transmission in the PLC of both sexes, but synaptic transmission is rescued by ketamine selectively in female mice. Thus, ketamine may have therapeutic potential as an ethanol binge suppressing agent selectively in female subjects.
Collapse
Affiliation(s)
- Nicole A Crowley
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Sarah N Magee
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA
| | - Mengyang Feng
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA; Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, 16802, USA; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Sarah J Jefferson
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Christian J Morris
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Nigel C Dao
- Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA; Department of Biobehavioral Health, Pennsylvania State University, University Park, PA, 16802, USA
| | - Dakota F Brockway
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA
| | - Bernhard Luscher
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA; Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, 16802, USA; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes for the Life Sciences, Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|
39
|
Ran Y, Jin Z, Chen X, Zhao N, Fang X, Zhang L, Zhang Y, Li Y. Hypidone Hydrochloride (YL-0919) Produces a Fast-Onset Reversal of the Behavioral and Synaptic Deficits Caused by Chronic Stress Exposure. Front Cell Neurosci 2018; 12:395. [PMID: 30524234 PMCID: PMC6256289 DOI: 10.3389/fncel.2018.00395] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 10/15/2018] [Indexed: 12/13/2022] Open
Abstract
Our previous study showed that hypidone hydrochloride (YL-0919), a partial serotonin 1A (5-HT1A) receptor agonist and 5-HT reuptake inhibitor, exerts a significant antidepressant effect in various animal models. The aim of the present study was to further investigate the underlying mechanisms and whether it could act as a fast-onset antidepressant. In the current study, depressive-like behavior was induced in rats by a chronic unpredictable stress (CUS) model and assessed with the Sucrose Preference Test (SPT). Treatment with YL-0919 (2.5 mg/kg, i.g.), but not with fluoxetine (Flx; 10 mg/kg, i.g.), caused a fast improvement in the SPT scores. In CUS-exposed rats, YL-0919 treatment for 5 days decreased the immobility time in a forced swimming test (FST), and a 10-day treatment decreased the latency to feed in a Novelty-Suppressed Feeding Test (NSFT). In addition to the behavioral tests, the effects of YL-0919 on synaptic protein expression were also evaluated. Western blotting showed that YL-0919 significantly enhanced the expression levels of synaptic proteins such as synapsin I, postsynaptic density protein 95 (PSD95), phosphorylated mammalian targeting of rapamycin (pmTOR) and brain-derived neurotrophic factor (BDNF) in the hippocampus. To determine how the mTOR signaling is involved in the fast-onset antidepressant-like effects of YL-0919, the mTOR-specific inhibitor rapamycin was administered intracerebroventricularly (i.c.v.) together with the YL-0919 treatment. The observed changes in behavioral tests and protein expression could be reversed by rapamycin treatment. This suggests that the fast-onset antidepressant effects of YL-0919 were partially caused by changes in synaptogenesis mediated by activation of mTOR pathways. Our data suggest that YL-0919 may be a powerful/effective antidepressant with fast-onset.
Collapse
Affiliation(s)
- Yuhua Ran
- State Key Laboratory of Toxicology Medical Countermeasures, Beijing Key Laboratories of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Academy of Military Sciences, Beijing, China
| | - Zengliang Jin
- State Key Laboratory of Toxicology Medical Countermeasures, Beijing Key Laboratories of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Academy of Military Sciences, Beijing, China
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaofei Chen
- State Key Laboratory of Toxicology Medical Countermeasures, Beijing Key Laboratories of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Academy of Military Sciences, Beijing, China
| | - Nan Zhao
- State Key Laboratory of Toxicology Medical Countermeasures, Beijing Key Laboratories of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Academy of Military Sciences, Beijing, China
| | - Xinxin Fang
- State Key Laboratory of Toxicology Medical Countermeasures, Beijing Key Laboratories of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Academy of Military Sciences, Beijing, China
| | - Liming Zhang
- State Key Laboratory of Toxicology Medical Countermeasures, Beijing Key Laboratories of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Academy of Military Sciences, Beijing, China
| | - Youzhi Zhang
- State Key Laboratory of Toxicology Medical Countermeasures, Beijing Key Laboratories of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Academy of Military Sciences, Beijing, China
| | - Yunfeng Li
- State Key Laboratory of Toxicology Medical Countermeasures, Beijing Key Laboratories of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Academy of Military Sciences, Beijing, China
| |
Collapse
|
40
|
Rajkumar R, Dawe GS. OBscure but not OBsolete: Perturbations of the frontal cortex in common between rodent olfactory bulbectomy model and major depression. J Chem Neuroanat 2018; 91:63-100. [DOI: 10.1016/j.jchemneu.2018.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/02/2018] [Accepted: 04/04/2018] [Indexed: 02/08/2023]
|
41
|
Dandekar MP, Peng T, McPherson DD, Quevedo J, Soares JC, Huang SL. Intravenous infusion of xenon-containing liposomes generates rapid antidepressant-like effects. Prog Neuropsychopharmacol Biol Psychiatry 2018; 86:140-149. [PMID: 29559371 DOI: 10.1016/j.pnpbp.2018.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/26/2018] [Accepted: 03/09/2018] [Indexed: 02/07/2023]
Abstract
AIM Similar to ketamine, xenon gas acts as a glutamatergic N-methyl-d-aspartate receptor antagonist, but devoid of propensity to cause untoward effects. Herein, we loaded xenon gas into a liposomal carrier called xenon-containing liposomes (Xe-liposome) for systemic delivery, and investigated its effect as an antidepressant and also analyzed synaptic biomarkers including brain-derived neurotrophic factor (BDNF), protein kinase B (AKT), mammalian target of rapamycin (mTOR), protein kinase C (PKC) and extracellular signal-regulated kinase-1/2 (ERK1/2) in blood and brain. METHODS Xe-liposomes (15 μl/mg) were prepared by a pressurized freeze-thaw method, and injected via the lateral tail vein (0.6 mL/rat) in male Wistar rats. The uncaging of xenon gas from circulating Xe-liposome was facilitated by continuous ultrasound application externally on the neck over the internal common carotid artery. One-hour after Xe-liposome infusion, animals were assessed for depression-like behaviors using a forced swimming test (FST), and spontaneous locomotor activity. Blood, as well as frontal cortex and hippocampal samples were obtained for immunoblotting and/or enzyme-linked immune sorbent assays. RESULTS Acute intravenous infusion of Xe-liposome, at 6 mg/kg, showed an increase in swimming time in the FST (p < 0.006), indicating antidepressant-like phenotypes. Higher doses of Xe-liposomes (9 mg/kg) failed to improve swimming duration. This behavioral discrepancy was not associated with locomotion aberrations, as gross activity of rats remained similar for both doses. In biochemical analyses of frontal cortex, protein levels of BDNF increased by 64%, and enhanced phosphorylation of AKT (43%) and mTOR (93%) was observed at the 6 mg/kg dose level of Xe-liposomes, while these biomarkers and phosphorylated PKC and ERK1/2 levels remained unchanged at the higher dose. Moreover, Xe-liposomal treatment did not change the plasma and protein levels of BDNF, and phosphorylated AKT, mTOR, PKC and ERK1/2 hippocampal expressions. CONCLUSION Xe-liposomes mediate a rapid antidepressant-like effect through activation of AKT/mTOR/BDNF signaling pathway.
Collapse
Affiliation(s)
- Manoj P Dandekar
- Department of Internal Medicine, Division of Cardiology, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, USA
| | - Tao Peng
- Department of Internal Medicine, Division of Cardiology, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, USA; Center for Clinical and Translational Sciences, The University of Texas Health Science Center at Houston, USA
| | - David D McPherson
- Department of Internal Medicine, Division of Cardiology, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, USA; Center for Clinical and Translational Sciences, The University of Texas Health Science Center at Houston, USA; Department of Biomedical Sciences, The University of Texas Medical School at Houston, USA; Memorial Hermann Heart and Vascular Institute-Texas Medical Center, USA
| | - Joao Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.; Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Jair C Soares
- Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, USA
| | - Shao-Ling Huang
- Department of Internal Medicine, Division of Cardiology, The University of Texas Health Science Center at Houston (UTHealth), McGovern Medical School, Houston, TX, USA; Center for Clinical and Translational Sciences, The University of Texas Health Science Center at Houston, USA.
| |
Collapse
|
42
|
Huang X, Wu H, Jiang R, Sun G, Shen J, Ma M, Ma C, Zhang S, Huang Z, Wu Q, Chen G, Tao W. The antidepressant effects of ɑ-tocopherol are related to activation of autophagy via the AMPK/mTOR pathway. Eur J Pharmacol 2018; 833:1-7. [DOI: 10.1016/j.ejphar.2018.05.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/28/2018] [Accepted: 05/17/2018] [Indexed: 12/25/2022]
|
43
|
Sun X, Li X, Pan R, Xu Y, Wang Q, Song M. Total Saikosaponins of Bupleurum yinchowense reduces depressive, anxiety-like behavior and increases synaptic proteins expression in chronic corticosterine-treated mice. Altern Ther Health Med 2018; 18:117. [PMID: 29609584 PMCID: PMC5879550 DOI: 10.1186/s12906-018-2186-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 03/22/2018] [Indexed: 12/22/2022]
Abstract
Background Bupleurum yinchowense Shan et Y. Li is widely used to treat depressive and anxiety disorders for hundreds of years in China. Total saikosaponins (TSS) is the major ingredient of Bupleurum yinchowense. A-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor and subsequent mammalian target of rapamycin (mTOR) signaling is responsible for synaptic maturation and may contribution to the synaptic alteration underlying depression. The aim of the study was to investigate the antidepressant-like and anxiolytic effect of TSS in chronic corticosterone-treated mice. The effect of TSS on synaptic proteins expression and AMPA receptor-mTOR signaling pathway alteration was also evaluated. Methods Dose-response effect of TSS (12.5, 25, 50 mg/kg) was investigated in forced swim test (FST) in ICR male mice. In the chronic corticosterine-treated model, TSS was given intragastrically once a day for 2 weeks and continued through the behavior testing period. Behavior tests and AMPA receptor related signaling pathway were investigated. Results TSS (25 and 50 mg/kg) decreased the immobility time in the FST when compared with the control group. TSS (25 mg/kg) showed antidepressant-like and anxiolytic effects in the chronic corticosterone treatment model in mice. TSS increased hippocampal synaptic proteins (synapsin-1 and postsynaptic density protein 95) expression. Immunohistochemistry analysis showed that TSS significantly increased the synapsin-1 expression in CA3 of hippocampus. TSS also increased hippocampal phosphorylation expression of GluR1 Ser 845 (AMPA receptor subunit) and its downstream regulators extracellular signaling-regulated kinase (ERK), protein kinase B (Akt) and mTOR. Conclusion TSS produces antidepressant-like and anxiolytic effects and increases synaptic proteins expression which may be mediated by induction of AMPA receptor and subsequent mTOR signaling pathway.
Collapse
|
44
|
Abstract
Traditional pharmacological treatments for depression have a delayed therapeutic onset, ranging from several weeks to months, and there is a high percentage of individuals who never respond to treatment. In contrast, ketamine produces rapid-onset antidepressant, anti-suicidal, and anti-anhedonic actions following a single administration to patients with depression. Proposed mechanisms of the antidepressant action of ketamine include N-methyl-D-aspartate receptor (NMDAR) modulation, gamma aminobutyric acid (GABA)-ergic interneuron disinhibition, and direct actions of its hydroxynorketamine (HNK) metabolites. Downstream actions include activation of the mechanistic target of rapamycin (mTOR), deactivation of glycogen synthase kinase-3 and eukaryotic elongation factor 2 (eEF2), enhanced brain-derived neurotrophic factor (BDNF) signaling, and activation of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors (AMPARs). These putative mechanisms of ketamine action are not mutually exclusive and may complement each other to induce potentiation of excitatory synapses in affective-regulating brain circuits, which results in amelioration of depression symptoms. We review these proposed mechanisms of ketamine action in the context of how such mechanisms are informing the development of novel putative rapid-acting antidepressant drugs. Such drugs that have undergone pre-clinical, and in some cases clinical, testing include the muscarinic acetylcholine receptor antagonist scopolamine, GluN2B-NMDAR antagonists (i.e., CP-101,606, MK-0657), (2R,6R)-HNK, NMDAR glycine site modulators (i.e., 4-chlorokynurenine, pro-drug of the glycineB NMDAR antagonist 7-chlorokynurenic acid), NMDAR agonists [i.e., GLYX-13 (rapastinel)], metabotropic glutamate receptor 2/3 (mGluR2/3) antagonists, GABAA receptor modulators, and drugs acting on various serotonin receptor subtypes. These ongoing studies suggest that the future acute treatment of depression will typically occur within hours, rather than months, of treatment initiation.
Collapse
Affiliation(s)
- Panos Zanos
- Department of Psychiatry, University of Maryland School of Medicine, Rm. 934F MSTF, 685 W. Baltimore St., Baltimore, MD, 21201, USA.
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Physiology, University of Maryland School of Medicine, St. BRB 5-007, 655 W. Baltimore St., Baltimore, MD, 21201, USA, Baltimore, MD, 21201, USA
| | - Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Todd D Gould
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Psychiatry, University of Maryland School of Medicine, Rm. 936 MSTF, 685 W. Baltimore St., Baltimore, MD, 21201, USA
| |
Collapse
|
45
|
McMurray KMJ, Ramaker MJ, Barkley-Levenson AM, Sidhu PS, Elkin P, Reddy MK, Guthrie ML, Cook JM, Rawal VH, Arnold LA, Dulawa SC, Palmer AA. Identification of a novel, fast-acting GABAergic antidepressant. Mol Psychiatry 2018; 23:384-391. [PMID: 28322281 PMCID: PMC5608625 DOI: 10.1038/mp.2017.14] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 12/07/2016] [Accepted: 01/09/2017] [Indexed: 01/04/2023]
Abstract
Current pharmacotherapies for depression exhibit slow onset, side effects and limited efficacy. Therefore, identification of novel fast-onset antidepressants is desirable. GLO1 is a ubiquitous cellular enzyme responsible for the detoxification of the glycolytic byproduct methylglyoxal (MG). We have previously shown that MG is a competitive partial agonist at GABA-A receptors. We examined the effects of genetic and pharmacological inhibition of GLO1 in two antidepressant assay models: the tail suspension test (TST) and the forced swim test (FST). We also examined the effects of GLO1 inhibition in three models of antidepressant onset: the chronic FST (cFST), chronic mild stress (CMS) paradigm and olfactory bulbectomy (OBX). Genetic knockdown of Glo1 or pharmacological inhibition using two structurally distinct GLO1 inhibitors (S-bromobenzylglutathione cyclopentyl diester (pBBG) or methyl-gerfelin (MeGFN)) reduced immobility in the TST and acute FST. Both GLO1 inhibitors also reduced immobility in the cFST after 5 days of treatment. In contrast, the serotonin reuptake inhibitor fluoxetine (FLX) reduced immobility after 14, but not 5 days of treatment. Furthermore, 5 days of treatment with either GLO1 inhibitor blocked the depression-like effects induced by CMS on the FST and coat state, and attenuated OBX-induced locomotor hyperactivity. Finally, 5 days of treatment with a GLO1 inhibitor (pBBG), but not FLX, induced molecular markers of the antidepressant response including brain-derived neurotrophic factor (BDNF) induction and increased phosphorylated cyclic-AMP response-binding protein (pCREB) to CREB ratio in the hippocampus and medial prefrontal cortex (mPFC). Our findings indicate that GLO1 inhibitors may provide a novel and fast-acting pharmacotherapy for depression.
Collapse
Affiliation(s)
- Katherine M. J. McMurray
- Committee on Neurobiology, University of Chicago, Chicago IL 60637, USA,Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Marcia J. Ramaker
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago IL 60637, USA
| | - Amanda M. Barkley-Levenson
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Preetpal S. Sidhu
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin – Milwaukee, Milwaukee, WI 53211, USA
| | - Pavel Elkin
- Department of Chemistry, University of Chicago, Chicago IL 60637, USA
| | - M. Kashi Reddy
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin – Milwaukee, Milwaukee, WI 53211, USA
| | - Margaret L. Guthrie
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin – Milwaukee, Milwaukee, WI 53211, USA
| | - James M. Cook
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin – Milwaukee, Milwaukee, WI 53211, USA
| | - Viresh H. Rawal
- Department of Chemistry, University of Chicago, Chicago IL 60637, USA
| | - Leggy A. Arnold
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin – Milwaukee, Milwaukee, WI 53211, USA
| | - Stephanie C. Dulawa
- Committee on Neurobiology, University of Chicago, Chicago IL 60637, USA,Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago IL 60637, USA,Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Abraham A. Palmer
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA,Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago IL 60637, USA,Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA,Institute for Genome Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA,Corresponding Author: Abraham A. Palmer, Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093,
| |
Collapse
|
46
|
An Interaction between Serotonin Receptor Signaling and Dopamine Enhances Goal-Directed Vigor and Persistence in Mice. J Neurosci 2018; 38:2149-2162. [PMID: 29367407 DOI: 10.1523/jneurosci.2088-17.2018] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 01/09/2018] [Accepted: 01/14/2018] [Indexed: 11/21/2022] Open
Abstract
The functionally selective 5-HT2C receptor ligand SB242084 can increase motivation and have rapid onset anti-depressant-like effects. We sought to identify the specific behavioral effects of SB242084 treatment and elucidate the mechanism in female and male mice. Using a quantitative behavioral approach, we determined that SB242084 increases the vigor and persistence of goal-directed activity across different types of physical work, particularly when work requirements are demanding. We found this influence of SB242084 on effort, rather than reward to be reflected in striatal DA measured during behavior. Using in vivo fast scan cyclic voltammetry, we found that SB242084 has no effect on reward-related phasic DA release in the NAc. Using in vivo microdialysis to measure tonic changes in extracellular DA, we also found no changes in the NAc. In contrast, SB242084 treatment increases extracellular DA in the dorsomedial striatum, an area that plays a key role in response vigor. These findings have several implications. At the behavioral level, this work shows that the capacity to work in demanding situations can be increased, without a generalized increase in motor activity or reward value. At the circuit level, we identified a pathway restricted potentiation of DA release and showed that this was the reason for the increased response vigor. At the cellular level, we show that a specific serotonin receptor cross talks to the DA system. Together, this information provides promise for the development of treatments for apathy, a serious clinical condition that can afflict patients with psychiatric and neurological disorders.SIGNIFICANCE STATEMENT Motivated behaviors are modulated by reward value, effort demands, and cost-benefit computations. This information drives the decision to act, which action to select, and the intensity with which the selected action is performed. Because these behavioral processes are all regulated by DA signaling, it is very difficult to influence selected aspects of motivated behavior without affecting others. Here we identify a pharmacological treatment that increases the vigor and persistence of responding in mice, without increasing generalized activity or changing reactions to rewards. We show that the 5-HT2C-selective ligand boosts motivation by potentiating activity-dependent DA release in the dorsomedial striatum. These results reveal a novel strategy for treating patients with motivational deficits, avolition, or apathy.
Collapse
|
47
|
Rodríguez-Landa JF, Cueto-Escobedo J, Flores-Aguilar LÁ, Rosas-Sánchez GU, Rovirosa-Hernández MDJ, García-Orduña F, Carro-Juárez M. The Aqueous Crude Extracts of Montanoa frutescens and Montanoa grandiflora Reduce Immobility Faster Than Fluoxetine Through GABA A Receptors in Rats Forced to Swim. J Evid Based Integr Med 2018; 23:2515690X18762953. [PMID: 29540064 PMCID: PMC5871057 DOI: 10.1177/2515690x18762953] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 02/07/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Montanoa frutescens and Montanoa grandiflora have been indistinctly used for centuries in traditional Mexican medicine for reproductive impairments, anxiety, and mood disorders. Preclinical studies support their aphrodisiac and anxiolytic properties, but their effects on mood are still unexplored. METHODS The effects of 25 and 50 mg/kg of M frutescens and M grandiflora extracts were evaluated on days 1, 7, 14, 21, and 28 of treatment, and compared with fluoxetine (1 mg/kg) and Remotiv (7.14 mg/kg) in Wistar rats. The participation of GABAA receptor in the effects produced by the treatments was explored. RESULTS Montanoa extracts reduced immobility since day 1 of treatment, while fluoxetine and Remotiv required 14 days. The GABAA antagonism blocked the effects of Montanoa extracts, but not of fluoxetine or Remotiv. CONCLUSIONS Montanoa extracts prevented quickly the stress-induced behaviors in the swimming test through action at the GABAA receptor, exerting a protective effect different to the typical antidepressants drugs.
Collapse
|
48
|
Luethi D, Hoener MC, Liechti ME. Effects of the new psychoactive substances diclofensine, diphenidine, and methoxphenidine on monoaminergic systems. Eur J Pharmacol 2018; 819:242-247. [DOI: 10.1016/j.ejphar.2017.12.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 12/31/2022]
|
49
|
Phillips BU, Dewan S, Nilsson SRO, Robbins TW, Heath CJ, Saksida LM, Bussey TJ, Alsiö J. Selective effects of 5-HT2C receptor modulation on performance of a novel valence-probe visual discrimination task and probabilistic reversal learning in mice. Psychopharmacology (Berl) 2018; 235:2101-2111. [PMID: 29682701 PMCID: PMC6015605 DOI: 10.1007/s00213-018-4907-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 04/12/2018] [Indexed: 12/14/2022]
Abstract
RATIONALE Dysregulation of the serotonin (5-HT) system is a pathophysiological component in major depressive disorder (MDD), a condition closely associated with abnormal emotional responsivity to positive and negative feedback. However, the precise mechanism through which 5-HT tone biases feedback responsivity remains unclear. 5-HT2C receptors (5-HT2CRs) are closely linked with aspects of depressive symptomatology, including abnormalities in reinforcement processes and response to stress. Thus, we aimed to determine the impact of 5-HT2CR function on response to feedback in biased reinforcement learning. METHODS We used two touchscreen assays designed to assess the impact of positive and negative feedback on probabilistic reinforcement in mice, including a novel valence-probe visual discrimination (VPVD) and a probabilistic reversal learning procedure (PRL). Systemic administration of a 5-HT2CR agonist and antagonist resulted in selective changes in the balance of feedback sensitivity bias on these tasks. RESULTS Specifically, on VPVD, SB 242084, the 5-HT2CR antagonist, impaired acquisition of a discrimination dependent on appropriate integration of positive and negative feedback. On PRL, SB 242084 at 1 mg/kg resulted in changes in behaviour consistent with reduced sensitivity to positive feedback. In contrast, WAY 163909, the 5-HT2CR agonist, resulted in changes associated with increased sensitivity to positive feedback and decreased sensitivity to negative feedback. CONCLUSIONS These results suggest that 5-HT2CRs tightly regulate feedback sensitivity bias in mice with consequent effects on learning and cognitive flexibility and specify a framework for the influence of 5-HT2CRs on sensitivity to reinforcement.
Collapse
Affiliation(s)
- Benjamin U. Phillips
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK
| | - Sigma Dewan
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK
| | - Simon R. O. Nilsson
- Rodent Behavioral Core, Department of Neuroscience and Physiology, Neuroscience Institute, New York University Medical Center, New York, NY 10016 USA
| | - Trevor W. Robbins
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK
| | - Christopher J. Heath
- School of Life, Health and Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA UK
| | - Lisa M. Saksida
- Molecular Medicine Research Group, Robarts Research Institute and Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON Canada ,The Brain and Mind Institute, Western University, London, ON Canada
| | - Timothy J. Bussey
- Molecular Medicine Research Group, Robarts Research Institute and Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON Canada ,The Brain and Mind Institute, Western University, London, ON Canada
| | - Johan Alsiö
- Department of Psychology and Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge, CB2 3EB UK
| |
Collapse
|
50
|
Morais M, Patrício P, Mateus-Pinheiro A, Alves ND, Machado-Santos AR, Correia JS, Pereira J, Pinto L, Sousa N, Bessa JM. The modulation of adult neuroplasticity is involved in the mood-improving actions of atypical antipsychotics in an animal model of depression. Transl Psychiatry 2017; 7:e1146. [PMID: 28585931 PMCID: PMC5537642 DOI: 10.1038/tp.2017.120] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 04/24/2017] [Accepted: 04/28/2017] [Indexed: 11/09/2022] Open
Abstract
Depression is a prevalent psychiatric disorder with an increasing impact in global public health. However, a large proportion of patients treated with currently available antidepressant drugs fail to achieve remission. Recently, antipsychotic drugs have received approval for the treatment of antidepressant-resistant forms of major depression. The modulation of adult neuroplasticity, namely hippocampal neurogenesis and neuronal remodeling, has been considered to have a key role in the therapeutic effects of antidepressants. However, the impact of antipsychotic drugs on these neuroplastic mechanisms remains largely unexplored. In this study, an unpredictable chronic mild stress protocol was used to induce a depressive-like phenotype in rats. In the last 3 weeks of stress exposure, animals were treated with two different antipsychotics: haloperidol (a classical antipsychotic) and clozapine (an atypical antipsychotic). We demonstrated that clozapine improved both measures of depressive-like behavior (behavior despair and anhedonia), whereas haloperidol aggravated learned helplessness in the forced-swimming test and behavior flexibility in a cognitive task. Importantly, an upregulation of adult neurogenesis and neuronal survival was observed in animals treated with clozapine, whereas haloperidol promoted a downregulation of these processes. Furthermore, clozapine was able to re-establish the stress-induced impairments in neuronal structure and gene expression in the hippocampus and prefrontal cortex. These results demonstrate the modulation of adult neuroplasticity by antipsychotics in an animal model of depression, revealing that the atypical antipsychotic drug clozapine reverts the behavioral effects of chronic stress by improving adult neurogenesis, cell survival and neuronal reorganization.
Collapse
Affiliation(s)
- M Morais
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal,ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - P Patrício
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal,ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - A Mateus-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal,ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - N D Alves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal,ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - A R Machado-Santos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal,ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J S Correia
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal,ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J Pereira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal,ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - L Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal,ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - N Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal,ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J M Bessa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal,ICVS/3B’s—PT Government Associate Laboratory, Braga/Guimarães, Portugal,Life and Health Science Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal. E-mail:
| |
Collapse
|