1
|
Marques SI, Sá SI, Carmo H, Carvalho F, Silva JP. Pharmaceutical-mediated neuroimmune modulation in psychiatric/psychological adverse events. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111114. [PMID: 39111563 DOI: 10.1016/j.pnpbp.2024.111114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/21/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024]
Abstract
The therapeutic use of many pharmaceuticals, including small molecules and biological therapies, has been associated with the onset of psychiatric and psychological adverse events (PPAEs), posing substantial concerns to patients' health and safety. These events, which encompass mood (e.g., depression, schizophrenia, suicidal ideation) and cognitive changes (e.g., learning and memory impairment, dementia) often remain undetected until advanced stages of clinical trials or pharmacovigilance, mostly because the mechanisms underlying the onset of PPAEs remain poorly understood. In recent years, the role of neuroimmune modulation (comprising an intricate interplay between various cell types and signaling pathways) in PPAEs has garnered substantial interest. Indeed, understanding these complex interactions would substantially contribute to increase the ability to predict the potential onset of PPAEs during preclinical stages of a new drug's R&D. This review provides a comprehensive summary of the most recent advances in neuroimmune modulation-related mechanisms contributing to the onset of PPAEs and their association with specific pharmaceuticals. Reported data strongly support an association between neuroimmune modulation and the onset of PPAEs. Pharmaceuticals may target specific molecular pathways and pathway elements (e.g., cholinergic and serotonergic systems), which in turn may directly or indirectly impact the inflammatory status and the homeostasis of the brain, regulating inflammation and neuronal function. Also, modulation of the peripheral immune system by pharmaceuticals that do not permeate the blood-brain barrier (e.g., monoclonal antibodies) may alter the neuroimmunomodulatory status of the brain, leading to PPAEs. In summary, this review underscores the diverse pathways through which drugs can influence brain inflammation, shedding light on potential targeted interventions.
Collapse
Affiliation(s)
- Sandra I Marques
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Susana I Sá
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| | - Helena Carmo
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Félix Carvalho
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - João P Silva
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
2
|
Padhan M, Mohapatra D, Mishra BR, Maiti R, Jena M. Efficacy and safety of add-on sarcosine in patients with major depressive disorder: A randomized controlled trial. J Psychiatr Res 2024; 178:298-304. [PMID: 39180989 DOI: 10.1016/j.jpsychires.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024]
Abstract
The main hurdles with current therapies for major depressive disorder (MDD) include lack of efficacy, therapeutic latency, and adverse drug reactions. Add-on therapy to conventional antidepressants may result in better therapeutic outcomes to overcome these obstacles. Sarcosine (N-methyl glycine), an endogenous amino acid that acts by modulating the NMDA receptor, is available as a dietary supplement. So, the present study was planned to evaluate the efficacy and safety of add-on sarcosine to SSRIs in MDD. In the present randomized, double-blind clinical trial (NCT04975100), 60 eligible participants with MDD were randomly assigned to either the test group (SSRI + sarcosine) or the control group (SSRI + placebo). Clinical and biochemical parameters like MADRS, CGI, serum BDNF, and serum glycine were assessed at baseline and eight weeks. The mean reduction in MADRS score was significant in both the control (-8.7, 95% CI: -11.0 to -6.4, p < 0.001) and the test group (-13.3, 95% CI: -14.9 to -11.7, p < 0.001), but the change in the test group was significantly greater (-4.6, 95% CI: -7.5 to -1.7, p = 0.003). The test group had a significantly higher response rate (p = 0.007) and remission rate (p = 0.038) compared to the control group. There was a significant increase in serum BDNF in both groups; however, the change in the test group was significantly higher than in the control group (p = 0.041). Similarly, the test group had a significantly higher increase in serum glycine than the control group (p < 0.001). Sarcosine may be considered an efficacious and safe add-on therapy to standard SSRIs in the management of MDD. ClinicalTrial.gov IdentifierNCT04975100.
Collapse
Affiliation(s)
- Milan Padhan
- Dept. of Pharmacology All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India.
| | - Debadatta Mohapatra
- Dept. of Psychiatry All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India.
| | - Biswa Ranjan Mishra
- Dept. of Psychiatry All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India.
| | - Rituparna Maiti
- Dept. of Pharmacology All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India.
| | - Monalisa Jena
- Dept. of Pharmacology All India Institute of Medical Sciences (AIIMS), Bhubaneswar, India.
| |
Collapse
|
3
|
Rodgers A, Bahceci D, Davey CG, Chatterton ML, Glozier N, Hopwood M, Loo C. Ensuring the affordable becomes accessible-lessons from ketamine, a new treatment for severe depression. Aust N Z J Psychiatry 2024; 58:109-116. [PMID: 37830221 DOI: 10.1177/00048674231203898] [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: 10/14/2023]
Abstract
In this paper, the case study of ketamine as a new treatment for severe depression is used to outline the challenges of repurposing established medicines and we suggest potential solutions. The antidepressant effects of generic racemic ketamine were identified over 20 years ago, but there were insufficient incentives for commercial entities to pursue its registration, or support for non-commercial entities to fill this gap. As a result, the evaluation of generic ketamine was delayed, piecemeal, uncoordinated, and insufficient to gain approval. Meanwhile, substantial commercial investment enabled the widespread registration of a patented, intranasal s-enantiomeric ketamine formulation (Spravato®) for depression. However, Spravato is priced at $600-$900/dose compared to ~$5/dose for generic ketamine, and the ~AUD$100 million annual government investment requested in Australia (to cover drug costs alone) has been rejected twice, leaving this treatment largely inaccessible for Australian patients 2 years after Therapeutic Goods Administration approval. Moreover, emerging evidence indicates that generic racemic ketamine is at least as effective as Spravato, but no comparative trials were required for regulatory approval and have not been conducted. Without action, this story will repeat regularly in the next decade with a new wave of psychedelic-assisted psychotherapy treatments, for which the original off-patent molecules could be available at low-cost and reduce the overall cost of treatment. Several systemic reforms are required to ensure that affordable, effective options become accessible; these include commercial incentives, public and public-private funding schemes, reduced regulatory barriers and more coordinated international public funding schemes to support translational research.
Collapse
Affiliation(s)
- Anthony Rodgers
- The George Institute for Global Health, University of New South Wales, Newtown, NSW, Australia
| | - Dilara Bahceci
- The George Institute for Global Health, University of New South Wales, Newtown, NSW, Australia
| | - Christopher G Davey
- Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| | - Mary Lou Chatterton
- Health Economics Group, Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Nick Glozier
- Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Malcolm Hopwood
- Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
| | - Colleen Loo
- School of Psychiatry, School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
- Black Dog Institute, Randwick, NSW, Australia
| |
Collapse
|
4
|
Bigio B, Azam S, Mathé AA, Nasca C. The neuropsychopharmacology of acetyl-L-carnitine (LAC): basic, translational and therapeutic implications. DISCOVER MENTAL HEALTH 2024; 4:2. [PMID: 38169018 PMCID: PMC10761640 DOI: 10.1007/s44192-023-00056-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024]
Abstract
Mitochondrial metabolism can contribute to nuclear histone acetylation among other epigenetic mechanisms. A central aspect of this signaling pathway is acetyl-L-carnitine (LAC), a pivotal mitochondrial metabolite best known for its role in fatty acid oxidation. Work from our and other groups suggested LAC as a novel epigenetic modulator of brain plasticity and a therapeutic target for clinical phenotypes of depression linked to childhood trauma. Aberrant mitochondrial metabolism of LAC has also been implicated in the pathophysiology of Alzheimer's disease. Furthermore, mitochondrial dysfunction is linked to other processes implicated in the pathophysiology of both major depressive disorders and Alzheimer's disease, such as oxidative stress, inflammation, and insulin resistance. In addition to the rapid epigenetic modulation of glutamatergic function, preclinical studies showed that boosting mitochondrial metabolism of LAC protects against oxidative stress, rapidly ameliorates insulin resistance, and reduces neuroinflammation by decreasing proinflammatory pathways such as NFkB in hippocampal and cortical neurons. These basic and translational neuroscience findings point to this mitochondrial signaling pathway as a potential target to identify novel mechanisms of brain plasticity and potential unique targets for therapeutic intervention targeted to specific clinical phenotypes.
Collapse
Affiliation(s)
- Benedetta Bigio
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
| | - Shofiul Azam
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Aleksander A Mathé
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Carla Nasca
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
5
|
Madjid N, Lidell V, Nordvall G, Lindskog M, Ögren SO, Forsell P, Sandin J. Antidepressant effects of novel positive allosteric modulators of Trk-receptor mediated signaling - a potential therapeutic concept? Psychopharmacology (Berl) 2023; 240:1789-1804. [PMID: 37394539 PMCID: PMC10349764 DOI: 10.1007/s00213-023-06410-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 06/20/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Major depressive disorder (MDD) is defined as a complex mental disorder which is characterized by a pervasive low mood and aversion to activity. Several types of neurotransmitter systems e.g. serotonergic, glutamatergic and noradrenergic systems have been suggested to play an important role in the origination of depression, but neurotrophins such as brain derived neurotrophic factor (BDNF) have also been implicated in the disease process. OBJECTIVES The purpose of this study was to examine the effects of a newly developed class of molecules, characterized as positive allosteric modulators of neurotrophin/Trk receptor mediated signaling (Trk-PAM), on neurotransmitter release and depression-like behavior in vivo. METHODS The effect of and possible interaction of neurotrophin/Trk signaling pathways with serotonergic and glutamatergic systems in the modulation of depression-related responses was studied using newly developed Trk-PAM compounds (ACD855, ACD856 and AC26845), as well as ketamine and fluoxetine in the forced swim test (FST) in rodents. Moreover, in vivo microdialysis in freely moving rats was used to assess changes in neurotransmitter levels in the rat. RESULTS The results from the study show that several different compounds, which all potentiate Trk-receptor mediated signaling, display antidepressant-like activity in the FST. Moreover, the data also indicate that the effects of both fluoxetine and ketamine in the FST, both used in clinical practice, are mediated via BDNF/TrkB signaling, which could have implications for novel therapies in MDD. CONCLUSIONS Trk-PAMs could provide an interesting avenue for the development of novel therapeutics in this area.
Collapse
Affiliation(s)
- Nather Madjid
- AlzeCure Pharma AB, Hälsovägen 7, 141 57, Huddinge, Sweden
| | | | - Gunnar Nordvall
- AlzeCure Pharma AB, Hälsovägen 7, 141 57, Huddinge, Sweden
- Division of Neuroscience, Care and Society, Department of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Maria Lindskog
- Division of Neuroscience, Care and Society, Department of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Sven-Ove Ögren
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Pontus Forsell
- AlzeCure Pharma AB, Hälsovägen 7, 141 57, Huddinge, Sweden
- Division of Neuroscience, Care and Society, Department of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Sandin
- AlzeCure Pharma AB, Hälsovägen 7, 141 57, Huddinge, Sweden.
- Division of Neuroscience, Care and Society, Department of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
6
|
Kristof Z, Gal Z, Torok D, Eszlari N, Sutori S, Erdelyi-Hamza B, Petschner P, Sperlagh B, Anderson IM, Deakin JFW, Bagdy G, Juhasz G, Gonda X. Variation along P2RX7 interacts with early traumas on severity of anxiety suggesting a role for neuroinflammation. Sci Rep 2023; 13:7757. [PMID: 37173368 PMCID: PMC10182087 DOI: 10.1038/s41598-023-34781-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 05/08/2023] [Indexed: 05/15/2023] Open
Abstract
Emotional stress is a leading risk factor in the development of neuropsychiatric disorders possibly via immune activation. P2X7 receptors promote neuroinflammation, and research suggests a relationship between chromosome region 12q2431, in which the P2X7R gene is located, and development of mood disorders, however, few studies concentrate on its association with anxiety. Our aim was to investigate the effects of P2RX7 variation in interaction with early childhood traumas and recent stressors on anxiety. 1752 participants completed questionnaires assessing childhood adversities and recent negative life events, provided data on anxiety using the Brief Symptom Inventory, and were genotyped for 681 SNPs in the P2RX7 gene, 335 of which passed quality control and were entered into linear regression models followed by a linkage disequilibrium-based clumping procedure to identify clumps of SNPs with a significant main or interaction effect. We identified a significant clump with top SNP rs67881993 and containing a set of 29SNPs that are in high LD, which significantly interacted with early childhood traumas but not with recent stress conveying a protective effect against increased anxiety in those exposed to early adversities. Our study demonstrated that P2RX7 variants interact with distal and more etiological stressors in influencing the severity of anxiety symptoms, supporting previous scarce results and demonstrating its role in moderating the effects of stress.
Collapse
Affiliation(s)
- Zsuliet Kristof
- Doctoral School of Mental Health Sciences, Semmelweis University, Budapest, Hungary
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Budapest, Hungary
- Department of Psychiatry and Psychotherapy, Semmelweis University, Gyulai Pál Str. 2, Budapest, 1085, Hungary
| | - Zsofia Gal
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Dora Torok
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Nora Eszlari
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Sara Sutori
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
| | - Berta Erdelyi-Hamza
- Doctoral School of Mental Health Sciences, Semmelweis University, Budapest, Hungary
- Department of Psychiatry and Psychotherapy, Semmelweis University, Gyulai Pál Str. 2, Budapest, 1085, Hungary
| | - Peter Petschner
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
- Bioinformatics Center, Institute of Chemical Research, Kyoto University, Uji, Kyoto, Japan
- Research Unit for Realization of Sustainable Society, Kyoto University, Uji, Kyoto, Japan
| | - Beata Sperlagh
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ian M Anderson
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biological, Medical and Human Sciences, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, UK
| | - John Francis William Deakin
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biological, Medical and Human Sciences, Manchester Academic Health Sciences Centre, The University of Manchester, Manchester, UK
| | - Gyorgy Bagdy
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Gabriella Juhasz
- Department of Pharmacodynamics, Faculty of Pharmacy, Semmelweis University, Budapest, Hungary
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Xenia Gonda
- Department of Psychiatry and Psychotherapy, Semmelweis University, Gyulai Pál Str. 2, Budapest, 1085, Hungary.
- NAP3.0-SE Neuropsychopharmacology Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
7
|
Gelin CF, Stenne B, Coate H, Hiscox A, Soyode-Johnson A, Wall JL, Lord B, Schoellerman J, Coe KJ, Wang K, Alcázar J, Chrovian CC, Dvorak CA, Carruthers NI, Koudriakova T, Balana B, Letavic MA. Discovery of a Series of Substituted 1 H-((1,2,3-Triazol-4-yl)methoxy)pyrimidines as Brain Penetrants and Potent GluN2B-Selective Negative Allosteric Modulators. J Med Chem 2023; 66:2877-2892. [PMID: 36757100 DOI: 10.1021/acs.jmedchem.2c01916] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Herein, we describe a series of substituted 1H-((1,2,3-triazol-4-yl)methoxy)pyrimidines as potent GluN2B negative allosteric modulators. Exploration of several five- and six-membered heterocycles led to the identification of O-linked pyrimidine analogues that possessed a balance of potency and desirable ADME profiles. Due to initial observations of metabolic saturation, early metabolite identification studies were conducted on compound 18, and the results drove further iterative optimization efforts to avoid the formation of undesired saturating metabolites. The comprehensive investigation of substitution on the pyrimidine moiety of the 1H-1,2,3-triazol-4-yl)methoxy)pyrimidines allowed for the identification of compound 31, which demonstrated high GluN2B receptor affinity, improved solubility, and a clean cardiovascular profile. Compound 31 was profiled in an ex vivo target engagement study in rats at a 10 mg/kg oral dose and achieved an ED50 of 1.7 mg/kg.
Collapse
Affiliation(s)
- Christine F Gelin
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Brice Stenne
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Heather Coate
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Afton Hiscox
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Akinola Soyode-Johnson
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Jessica L Wall
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Brian Lord
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Jeffrey Schoellerman
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Kevin J Coe
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Kai Wang
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Jesus Alcázar
- Janssen Research & Development, Janssen-Cilag, S.A., Jarama 75A, 45007 Toledo, Spain
| | - Christa C Chrovian
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Curt A Dvorak
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Nicholas I Carruthers
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Tatiana Koudriakova
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Bartosz Balana
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| | - Michael A Letavic
- Janssen Research & Development, 3210 Merryfield Row, San Diego, California 92121-1126, United States
| |
Collapse
|
8
|
Saleem U, Iman S, Ahmad B, Shah MA, Bibi S, Alqarni M, Khan MS, Shah GM, Khan H, Alhasani RH, Althobaiti NA, Albalawi AE. Antidepressant activity of phytochemicals of Mangifera indica seeds assisted by integrated computational analysis. Metab Brain Dis 2023; 38:483-505. [PMID: 35344129 DOI: 10.1007/s11011-022-00955-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/09/2022] [Indexed: 01/25/2023]
Abstract
Mangifera indica L., also known as mango, is a tropical fruit that belongs to the Anacardiaceae family and is prized for its juiciness, unique flavour, and worldwide popularity. The current study aimed to probe into antidepressant power (ADP) of MIS in animals and confirmation of ADP with in silico induced-fit molecular docking. The depression model was prepared by exposing mice to various stressors from 9:00 am to 2:00 pm during 42 days study period. MIS extract and fluoxetine were given daily for 30 min before exposing animals to stressors. ADP was evaluated by various behavioural tests and biochemical analysis. Results showed increased physical activity in mice under behavioural tests, plasma nitrite and malondialdehyde (MDA) levels and monoamine oxidase A (MAO-A) activity decreased dose-dependently in MIS treated mice and superoxide dismutases (SOD) levels increased in treated groups as compared to disease control. With the peculiar behaviour and significant interactions of the functional residues of target proteins with selected ligands along with the best absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties, it is concluded that catechin could be the best MAO-A inhibitor at a binding energy of -8.85 kcal/mol, and two hydrogen bonds were generated with Cys406 (A) and Gly443 (A) residues of the active binding site of MAO-A enzyme. While catechin at -6.86 kcal/mol generated three hydrogen bonds with Ala263 (A) and Gly434 (A) residues of the active site of monoamine oxidase B (MAO-B) enzyme and stabilized the best conformation. Therefore, it is highly recommended to test the selected lead-like compound catechin in the laboratory with biological system analysis to confirm its activity as MAO-A and MAO-B inhibitors so it can be declared as one of the novel therapeutic options with anti-depressant activity. Our findings concluded that M. indica seeds could be a significant and alternative anti-depressant therapy.
Collapse
Affiliation(s)
- Uzma Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, 38000, Pakistan.
| | - Shafa Iman
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Bashir Ahmad
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore, Pakistan
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, 38000, Pakistan.
- Department of Pharmacy, Hazara University, Mansehra, Pakistan.
| | - Shabana Bibi
- Yunnan Herbal Laboratory, College of Ecology and Environmental Sciences, Yunnan University, Kunming, 650091, Yunnan, China
- International Joint Research Center for Sustainable Utilization of Cordyceps Bioresources in China and South-East Asia, Yunnan University, Kunming, 650091, Yunnan, China
| | - Mohammed Alqarni
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, 21944, Saudi Arabia
| | - Muhammad Saad Khan
- Department of Biosciences, Faculty of Sciences, COMSATS University Islamabad, Sahiwal, Pakistan
| | - Ghulam Mujtaba Shah
- Department of Pharmacy, Hazara University, Mansehra, Pakistan
- Department of Botany, Hazara University, Mansehra, Pakistan
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Reem Hasaballah Alhasani
- Department of Biology, Faculty of Applied Science, Umm Al-Qura University, Makkah, 21961, Saudi Arabia
| | - Norah A Althobaiti
- Department of Biology, College of Science and Humanities, Shaqra University, Al-Quwaiiyah, Saudi Arabia
| | - Aishah E Albalawi
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| |
Collapse
|
9
|
Halaris A, Cook J. The Glutamatergic System in Treatment-Resistant Depression and Comparative Effectiveness of Ketamine and Esketamine: Role of Inflammation? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:487-512. [PMID: 36949323 DOI: 10.1007/978-981-19-7376-5_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
The glutamatergic system is the primary excitatory pathway within the CNS and is responsible for cognition, memory, learning, emotion, and mood. Because of its significant importance in widespread nervous system function, it is tightly regulated through multiple mechanisms, such as glutamate recycling, microglial interactions, and inflammatory pathways. Imbalance within the glutamatergic system has been implicated in a wide range of pathological conditions including neurodegenerative conditions, neuromuscular conditions, and mood disorders including depression. Major depressive disorder (MDD) is the most common mood disorder worldwide, has a high prevalence rate, and afflicts approximately 280 million people. While there are numerous treatments for the disease, 30-40% of patients are unresponsive to treatment and deemed treatment resistant; approximately another third experience only partial improvement (World Health Organization, Depression fact sheet [Internet], 2020). Esketamine, the S-enantiomer of ketamine, was approved by the Food and Drug Administration for treatment-resistant depression (TRD) in 2019 and has offered new hope to patients. It is the first treatment targeting the glutamatergic system through a complex mechanism. Numerous studies have implicated imbalance in the glutamatergic system in depression and treatment resistance. Esketamine and ketamine principally work through inhibition of the NMDA receptor, though more recent studies have implicated numerous other mechanisms mediating the antidepressant efficacy of these agents. These mechanisms include increase in brain-derived neurotrophic factor (BDNF), activation of mammalian target of the rapamycin complex (mTORC), and reduction in inflammation. Esketamine and ketamine have been shown to decrease inflammation in numerous ways principally through reducing pro-inflammatory cytokines (e.g., TNF-α, IL-6) (Loix et al., Acta Anaesthesiol Belg 62(1):47-58, 2011; Chen et al., Psychiatry Res 269:207-11, 2018; Kopra et al., J Psychopharmacol 35(8):934-45, 2021). This anti-inflammatory effect has also been shown to be involved in the antidepressive properties of both ketamine and esketamine (Chen et al., Psychiatry Res 269:207-11, 2018; Kopra et al., J Psychopharmacol 35(8):934-45, 2021).
Collapse
Affiliation(s)
- Angelos Halaris
- Department of Psychiatry, Loyola University Stritch School of Medicine, Maywood, IL, USA.
| | - John Cook
- Department of Psychiatry, Loyola University Stritch School of Medicine, Maywood, IL, USA
| |
Collapse
|
10
|
Adjunctive dopaminergic enhancement of esketamine in treatment-resistant depression. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110603. [PMID: 35842074 DOI: 10.1016/j.pnpbp.2022.110603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/10/2022] [Indexed: 11/22/2022]
Abstract
Esketamine is a novel treatment for treatment resistant depression (TRD) and was approved by the FDA in early 2019. It antagonizes the NMDA receptor providing rapid improvement in symptoms with a complex mechanism of action primarily mediated through glutamatergic activation. Significant barriers exist to widespread use of esketamine including durability of response, particularly in the maintenance phase. Since it must be administered in combination with an oral antidepressant, investigating appropriate treatments to be given concomitantly may further improve outcomes and response duration. Specifically, due to dysfunction in dopaminergic pathways in many patients with MDD and TRD, addition of a prodopaminergic agent, such as bupropion, may provide additional benefit and durability of response. Historically, the addition of a dopaminergic agent to traditional treatment (e.g., SSRI, SNRI) has been shown to improve response in TRD. While we have anecdotal evidence to support adjunctive dopaminergic enhancement of esketamine response, robust data are limited. There are case reports that exhibit efficacy with the use of a MAO-I in combination with ketamine supporting at least in part a dopaminergic component. Additionally, there is mechanistic rationale for the use of dopaminergic agents with a NMDA antagonist. This includes co-localization of NMDA and dopamine receptors as well as increased glutamatergic signaling due to dopamine-induced phosphorylation of AMPAR. Recently, AXS-05, an oral combination of dextromethorphan and bupropion, has shown promise in both MDD and TRD clinical trials highlighting the potential validity of this mechanism. This paper describes how dopaminergic enhancement may increase efficacy and durability of response with esketamine, encouraging further research into this treatment option.
Collapse
|
11
|
Sani G, Kotzalidis GD, Fiaschè F, Manfredi G, Ghaemi SN. Second messengers and their importance for novel drug treatments of patients with bipolar disorder. Int Rev Psychiatry 2022; 34:736-752. [PMID: 36786113 DOI: 10.1080/09540261.2022.2119073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Second messenger systems, like the cyclic nucleotide, glycogen synthase kinase-3β, phosphoinositide, and arachidonic acid cascades, are involved in bipolar disorder (BD). We investigated their role on the development of novel therapeutic drugs using second messenger mechanisms. PubMed search and narrative review. We used all relevant keywords for each second messenger cascade combining it with BD and related terms and combined all with novel/innovative treatments/drugs. Our search produced 31 papers most were reviews, and focussed on the PI3K/AKT-GSK-3β/Nrf2-NF-ĸB pathways. Only two human randomized clinical trials were identified, of ebselen, an antioxidant, and celecoxib, a cyclooxygenase-2 inhibitor, both with poor unsatisfactory results. Despite the fact that all second messenger systems are involved in the pathophysiology of BD, there are few experiments with novel drugs using these mechanisms. These mechanisms are a neglected and potentially major opportunity to transform the treatment of bipolar illness.
Collapse
Affiliation(s)
- Gabriele Sani
- Department of Neuroscience, Section of Psychiatry, Università Cattolica del Sacro Cuore, Rome, Italy.,Department of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Georgios D Kotzalidis
- Centro Lucio Bini, Rome, Italy.,NESMOS Department, Faculty of Medicine and Psychology, Sapienza University, Sant'Andrea University Hospital, Rome, Italy
| | - Federica Fiaschè
- NESMOS Department, Faculty of Medicine and Psychology, Sapienza University, Sant'Andrea University Hospital, Rome, Italy.,ASL Rieti, Servizio Psichiatrico Diagnosi e Cura, Ospedale San Camillo de Lellis, Rieti, Italy
| | - Giovanni Manfredi
- Centro Lucio Bini, Rome, Italy.,NESMOS Department, Faculty of Medicine and Psychology, Sapienza University, Sant'Andrea University Hospital, Rome, Italy
| | - S Nassir Ghaemi
- School of Medicine, Tufts University, Boston, MA, USA.,Lecturer on Psychiatry, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Efficacy of safinamide as add-on therapy after subthalamic nucleus deep brain stimulation in Parkinson disease. Neurol Sci 2022; 43:3187-3193. [DOI: 10.1007/s10072-021-05730-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 11/09/2021] [Indexed: 11/26/2022]
|
13
|
Stein CA, Penn A, Van Hope S, Dorsen CG, Mangini M. The Pharmacology and Clinical Applications of Psychedelic Medicines Within Midwifery Practice. J Midwifery Womens Health 2022; 67:373-383. [PMID: 35522087 PMCID: PMC9322528 DOI: 10.1111/jmwh.13371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 04/04/2022] [Accepted: 04/07/2022] [Indexed: 11/28/2022]
Abstract
The research and use of psychedelic medicines to treat common mental health disorders has increased substantially in the past 2 decades. At the same time, knowledge is relatively uncommon among midwives regarding (1) the relative benefits of psychedelic-assisted therapy, (2) best practices associated with the delivery of psychedelic-assisted therapy, and (3) responsible integration of this potentially useful intervention into mental health treatment plans. The purpose of this review is to describe current applications of psychedelic medicines to treat common mental health disorders, to describe the current legal status of these medicines used in this context, and to explore the potential for midwifery practice in this area with further training. This article also addresses the disparities regarding LGBTQIA+ and BIPOC populations in relation to this topic and their historical exclusion from research and treatment access in this field.
Collapse
Affiliation(s)
- Cindy A Stein
- California State University Monterey Bay School of Nursing, Seaside, California
| | - Andrew Penn
- University of California at San Francisco School of Nursing, San Francisco, California
| | | | - Caroline G Dorsen
- Division of Advanced Nursing Practice, Rutgers School of Nursing, Newark, New Jersey
| | - Mariavittoria Mangini
- Integral and Transpersonal Psychology Department, California Institute of Integral Studies, San Francisco, California
| |
Collapse
|
14
|
Liu T, Deng K, Xue Y, Yang R, Yang R, Gong Z, Tang M. Carnitine and Depression. Front Nutr 2022; 9:853058. [PMID: 35369081 PMCID: PMC8964433 DOI: 10.3389/fnut.2022.853058] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/11/2022] [Indexed: 11/13/2022] Open
Abstract
Depression has become one of the most common mental diseases in the world, but the understanding of its pathogenesis, diagnosis and treatments remains insufficient. Carnitine is a natural substance that exists in organisms, which can be synthesized in vivo or supplemented by intake. Relationships of carnitine with depression, bipolar disorder and other mental diseases have been reported in different studies. Several studies show that the level of acylcarnitines (ACs) changes significantly in patients with depression compared with healthy controls while the supplementation of acetyl-L-carnitine is beneficial to the treatment of depression. In this review, we aimed to clarify the effects of ACs in depressive patients and to explore whether ACs might be the biomarkers for the diagnosis of depression and provide new ideas to treat depression.
Collapse
Affiliation(s)
- Ting Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Institute for Rational and Safe Medication Practices, Central South University, Changsha, China
| | - Kunhong Deng
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ying Xue
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Institute for Rational and Safe Medication Practices, Central South University, Changsha, China
| | - Rui Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Institute for Rational and Safe Medication Practices, Central South University, Changsha, China
| | - Rong Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Institute for Rational and Safe Medication Practices, Central South University, Changsha, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Institute for Rational and Safe Medication Practices, Central South University, Changsha, China
| | - Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Institute for Rational and Safe Medication Practices, Central South University, Changsha, China
| |
Collapse
|
15
|
Frank D, Gruenbaum BF, Shelef I, Zvenigorodsky V, Severynovska O, Gal R, Dubilet M, Zlotnik A, Kofman O, Boyko M. Blood Glutamate Scavenging With Pyruvate as a Novel Preventative and Therapeutic Approach for Depressive-Like Behavior Following Traumatic Brain Injury in a Rat Model. Front Neurosci 2022; 16:832478. [PMID: 35237125 PMCID: PMC8883046 DOI: 10.3389/fnins.2022.832478] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/07/2022] [Indexed: 12/14/2022] Open
Abstract
Depression is a common and serious complication following traumatic brain injury (TBI). Both depression and TBI have independently been associated with pathologically elevated extracellular brain glutamate levels. In the setting of TBI, blood glutamate scavenging with pyruvate has been widely shown as an effective method to provide neuroprotection by reducing blood glutamate and subsequent brain glutamate levels. Here we evaluate pyruvate as a novel approach in the treatment and prevention of post-TBI depression-like behavior in a rat model. Rats were divided into five groups: (1) sham-operated control with pyruvate, (2) sham-operated control with placebo, (3) post-TBI with placebo, (4) post-TBI given preventative pyruvate, and (5) post-TBI treated with pyruvate. These groups had an equal number of females and males. Rats were assessed for depressive-like behavior, neurological status, and glutamate levels in the blood and brain. Post-TBI neurological deficits with concurrent elevations in glutamate levels were demonstrated, with peak glutamate levels 24 h after TBI. Following TBI, the administration of either prophylactic or therapeutic pyruvate led to reduced glutamate levels, improved neurologic recovery, and improved depressive-like behavior. Glutamate scavenging with pyruvate may be an effective prophylactic and therapeutic option for post-TBI depression by reducing associated elevations in brain glutamate levels.
Collapse
Affiliation(s)
- Dmitry Frank
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Benjamin F. Gruenbaum
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Ilan Shelef
- Department of Radiology, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Vladislav Zvenigorodsky
- Department of Radiology, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Olena Severynovska
- Department of Physiology, Faculty of Biology, Ecology and Medicine, Dnepropetrovsk State University, Dnepropetrovsk, Ukraine
| | - Ron Gal
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Michael Dubilet
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Alexander Zlotnik
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Ora Kofman
- Department of Psychology, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Matthew Boyko
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
- *Correspondence: Matthew Boyko,
| |
Collapse
|
16
|
Di Iorio R, Rossi S, Rossini PM. One century of healing currents into the brain from the scalp: From electroconvulsive therapy to repetitive transcranial magnetic stimulation for neuropsychiatric disorders. Clin Neurophysiol 2021; 133:145-151. [PMID: 34864511 DOI: 10.1016/j.clinph.2021.10.014] [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: 06/17/2021] [Revised: 10/01/2021] [Accepted: 10/30/2021] [Indexed: 11/27/2022]
Abstract
Electroconvulsive therapy (ECT) was applied for the first time in humans in 1938: after 80 years, it remains conceptually similar today except for modifications of the original protocol aimed to reduce adverse effects (as persistent memory deficits) without losing clinical efficacy. We illustrate the stages of development as well as ups and downs of ECT use in the last eighty years, and the impact that it still maintains for treatment of certain psychiatric conditions. Targeted, individualized and safe noninvasive neuromodulatory interventions are now possible for many neuropsychiatric disorders thanks to repetitive transcranial magnetic stimulation (rTMS) that injects currents in the brain through electromagnetic induction, powerful enough to depolarize cortical neurons and related networks. Although ECT and rTMS differ in basic concepts, mechanisms, tolerability, side effects and acceptability, and beyond their conceptual remoteness (ECT) or proximity (rTMS) to "precision medicine" approaches, the two brain stimulation techniques may be considered as complementary rather than competing in the current treatment of certain neuropsychiatric disorders.
Collapse
Affiliation(s)
- Riccardo Di Iorio
- Neurology Unit, Policlinic A. Gemelli Foundation IRCCS, Rome, Italy.
| | - Simone Rossi
- Siena Brain Investigation and Neuromodulation Lab (Si-BIN Lab), Department of Medicine, Surgery and Neuroscience, Section of Neurology and Clinical Neurophysiology, Policlinico Le Scotte, University of Siena, Italy
| | - Paolo M Rossini
- Department of Neuroscience & Neurorehabilitation, IRCCS San Raffaele-Pisana, Rome, Italy
| |
Collapse
|
17
|
Emerging role of microRNAs as novel targets of antidepressants. Asian J Psychiatr 2021; 66:102906. [PMID: 34740127 DOI: 10.1016/j.ajp.2021.102906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/20/2021] [Indexed: 12/31/2022]
|
18
|
Wang YT, Wang XL, Feng ST, Chen NH, Wang ZZ, Zhang Y. Novel rapid-acting glutamatergic modulators: Targeting the synaptic plasticity in depression. Pharmacol Res 2021; 171:105761. [PMID: 34242798 DOI: 10.1016/j.phrs.2021.105761] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/04/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
Major depressive disorder (MDD) is severely prevalent, and conventional monoaminergic antidepressants gradually exhibit low therapeutic efficiency, especially for patients with treatment-resistant depression. A neuroplasticity hypothesis is an emerging advancement in the mechanism of depression, mainly expressed in the glutamate system, e.g., glutamate receptors and signaling. Dysfunctional glutamatergic neurotransmission is currently considered to be closely associated with the pathophysiology of MDD. Biological function, pharmacological action, and signal attributes in the glutamate system both regulate the neural process. Specific functional subunits could be therapeutic targets to explore the novel glutamatergic modulators, which have fast-acting, and relatively sustained antidepressant effects. Here, the present review summarizes the pathophysiology of MDD found in the glutamate system, exploring the role of glutamate receptors and their downstream effects. These convergent mechanisms have prompted the development of other modulators targeting on glutamate system, including N-methyl-d-aspartate receptor antagonists, selective GluN2B-specific antagonists, glycine binding site agents, and regulators of metabotropic glutamate receptors. Relevant researches underly the putative mechanisms of these drugs, which reverse the damage of depression by regulating glutamatergic neurotransmission. It also provides further insight into the mechanism of depression and exploring potential targets for novel agent development.
Collapse
Affiliation(s)
- Ya-Ting Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiao-Le Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Si-Tong Feng
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
19
|
Impact of SAfinamide on Depressive Symptoms in Parkinson's Disease Patients (SADness-PD Study): A Multicenter Retrospective Study. Brain Sci 2021; 11:brainsci11020232. [PMID: 33668408 PMCID: PMC7918129 DOI: 10.3390/brainsci11020232] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/24/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND We aimed to assess the effects of safinamide on depression, motor symptoms, and the serotonin syndrome related to its co-administration with antidepressants in patients with Parkinson's disease (PD). METHODS We retrospectively analyzed the data of patients at 1 and 3 months of follow-up compared to baseline. RESULTS n = 82 (safinamide 50 mg = 22, 100 mg = 60, with antidepressants = 44). First, we found improvement in depression (Hamilton Depression Rating Scale: -6 ± 5.10 at 1 month and -7.27 ± 5.10 at 3 months, p < 0.0001; Patient Global Impression of Improvement Scale: 60.3% and 69.5% of patients at 1 and 3 months reported some improvement). Second, safinamide improved the daily life activities and motor symptoms/motor complications (Unified Parkinson's Disease Rating Scale (UPDRS-II): -2.51 ± 6.30 and -2.47 ± 6.11 at 1 and 3 months, p < 0.0001; III: -3.58 ± 8.68 and -4.03 ± 8.95 at 1 and 3 months, p < 0.0001; IV: -0.61 ± 2.61 and -0.8 ± 2.53 at 1 and 3 months, p < 0.0001). Third, 7.31% and 8.53% of patients developed non-severe adverse events related to safinamide at 1 and 3 months. Serotonin syndrome was not observed in the patients treated with antidepressants; some isolated serotonin syndrome symptoms were reported. CONCLUSIONS Safinamide could be useful for treating depression in PD; it was effective for motor symptoms and motor complications and safe even when co-administered with antidepressants.
Collapse
|
20
|
Sarmah T, Bhattacharyya DK. A study of tools for differential co-expression analysis for RNA-Seq data. INFORMATICS IN MEDICINE UNLOCKED 2021. [DOI: 10.1016/j.imu.2021.100740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
21
|
Sonmez AI, Lewis CP, Port JD, Cabello-Arreola A, Blacker CJ, Seewoo BJ, McKean AJ, Leffler JM, Frye MA, Croarkin PE. Glutamatergic Correlates of Bipolar Symptoms in Adolescents. J Child Adolesc Psychopharmacol 2020; 30:599-605. [PMID: 33179961 PMCID: PMC7757593 DOI: 10.1089/cap.2020.0082] [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: 12/20/2022]
Abstract
Objectives: Prior studies demonstrate elevated cortical glutamate (Glu) in patients with bipolar disorder (BD). Studies assessing neurochemistry in early stages of bipolar illness before the emergence of manic symptoms are lacking. This study aimed to examine neurochemical correlates measured by proton magnetic resonance spectroscopy (1H-MRS) and a dimensional measure of bipolarity in a sample of depressed adolescents. Methods: Adolescent participants (aged 13-21 years) underwent a semistructured diagnostic interview and clinical assessment, which included the General Behavior Inventory Parent Version (P-GBI), a 73-item, parent-rated assessment of symptoms and behaviors. 1H-MRS scans of a left dorsolateral prefrontal cortex (L-DLPFC) voxel (8 cm3) were collected using a two-dimensional J-averaged sequence to assess N-acetylaspartate (NAA), Glu, Glx (glutamate + glutamine), and NAA/Glx concentrations. We used generalized linear models to assess the relationships between P-GBI scores and metabolite levels in L-DLPFC. Results: Thirty-six participants (17 healthy controls, 19 depressed) underwent 1H-MRS scans and clinical evaluation with the P-GBI. There was a significant negative relationship between P-GBI score and L-DLPFC NAA/Glx in the whole sample. However, the magnitude of the effect was small and statistical significance was lost after correcting for multiple comparisons. Conclusions: These preliminary results suggest that NAA/Glx may have utility as a marker of bipolar traits in healthy and depressed adolescents. If replicated, 1H-MRS measures of glutamatergic metabolism anomalies might have a role in identifying depressed adolescents at risk for mixed symptom presentations or BD. Identifying bipolarity in the early stages of the disease would have a significant impact on treatment planning and prognosis. Further longitudinal studies should examine neurochemical correlates of mood state during the developmental emergence of BD.
Collapse
Affiliation(s)
- A. Irem Sonmez
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA.,Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Charles P. Lewis
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - John D. Port
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA.,Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Caren J. Blacker
- Department of Psychiatry and Behavioral Sciences, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Bhedita J. Seewoo
- Experimental and Regenerative Neurosciences, School of Biological Sciences, University of Western Australia, Crawley, Australia
| | - Alastair J. McKean
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jarrod M. Leffler
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark A. Frye
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA
| | - Paul E. Croarkin
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota, USA.,Address correspondence to: Paul E. Croarkin, DO, MSCS, Department of Psychiatry and Psychology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
22
|
Neis VB, Moretti M, Rosa PB, Dalsenter YDO, Werle I, Platt N, Kaufmann FN, Rosado AF, Besen MH, Rodrigues ALS. The involvement of PI3K/Akt/mTOR/GSK3β signaling pathways in the antidepressant-like effect of AZD6765. Pharmacol Biochem Behav 2020; 198:173020. [DOI: 10.1016/j.pbb.2020.173020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/10/2020] [Accepted: 08/26/2020] [Indexed: 12/31/2022]
|
23
|
Yalin N, Young AH. Pharmacological Treatment of Bipolar Depression: What are the Current and Emerging Options? Neuropsychiatr Dis Treat 2020; 16:1459-1472. [PMID: 32606699 PMCID: PMC7294105 DOI: 10.2147/ndt.s245166] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/15/2020] [Indexed: 12/15/2022] Open
Abstract
Depression accounts for the predominant burden associated with bipolar disorder. The identification and management of bipolar depression are challenging, since bipolar depression differs from unipolar depression, responding poorly to traditional antidepressants, which may also induce a switch to hypomania/mania, mixed states and/or cause rapid cycling. Current treatment options for bipolar depression are limited and guidelines vary greatly in their recommendations, reflecting gaps and inconsistencies in the current evidence base. Moreover, some treatment options, such as quetiapine and olanzapine-fluoxetine, although clearly efficacious, may be associated with adverse cardiometabolic side effects, which can be detrimental to the long-term physical health and well-being of patients, increasing the likelihood of treatment non-adherence and relapse. Evidence for some more recent therapeutic options, including lurasidone and cariprazine, suggests that patients' symptoms can be effectively managed without compromising their physical health. In addition, novel agents targeting alternative neurotransmitter pathways and inflammatory processes (such as ketamine and N-acetyl cysteine) are emerging as promising potential options for the treatment of bipolar depression in the future.
Collapse
Affiliation(s)
- Nefize Yalin
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Allan H Young
- Centre for Affective Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| |
Collapse
|
24
|
Andronis C, Silva JP, Lekka E, Virvilis V, Carmo H, Bampali K, Ernst M, Hu Y, Loryan I, Richard J, Carvalho F, Savić MM. Molecular basis of mood and cognitive adverse events elucidated via a combination of pharmacovigilance data mining and functional enrichment analysis. Arch Toxicol 2020; 94:2829-2845. [PMID: 32504122 PMCID: PMC7395038 DOI: 10.1007/s00204-020-02788-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/20/2020] [Indexed: 01/04/2023]
Abstract
Drug-induced Mood- and Cognition-related adverse events (MCAEs) are often only detected during the clinical trial phases of drug development, or even after marketing, thus posing a major safety concern and a challenge for both pharmaceutical companies and clinicians. To fill some gaps in the understanding and elucidate potential biological mechanisms of action frequently associated with MCAEs, we present a unique workflow linking observational population data with the available knowledge at molecular, cellular, and psychopharmacology levels. It is based on statistical analysis of pharmacovigilance reports and subsequent signaling pathway analyses, followed by evidence-based expert manual curation of the outcomes. Our analysis: (a) ranked pharmaceuticals with high occurrence of such adverse events (AEs), based on disproportionality analysis of the FDA Adverse Event Reporting System (FAERS) database, and (b) identified 120 associated genes and common pathway nodes possibly underlying MCAEs. Nearly two-thirds of the identified genes were related to immune modulation, which supports the critical involvement of immune cells and their responses in the regulation of the central nervous system function. This finding also means that pharmaceuticals with a negligible central nervous system exposure may induce MCAEs through dysregulation of the peripheral immune system. Knowledge gained through this workflow unravels putative hallmark biological targets and mediators of drug-induced mood and cognitive disorders that need to be further assessed and validated in experimental models. Thereafter, they can be used to substantially improve in silico/in vitro/in vivo tools for predicting these adversities at a preclinical stage.
Collapse
Affiliation(s)
| | - João Pedro Silva
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | | | | | - Helena Carmo
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Konstantina Bampali
- Department of Molecular Neurosciences, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Margot Ernst
- Department of Molecular Neurosciences, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Yang Hu
- Translational PKPD Group, Department of Pharmaceutical Biosciences, Associate Member of SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Irena Loryan
- Translational PKPD Group, Department of Pharmaceutical Biosciences, Associate Member of SciLifeLab, Uppsala University, Uppsala, Sweden
| | - Jacques Richard
- Sanofi R&D, 371 avenue Professeur Blayac, 34000, Montpellier, France
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| | - Miroslav M Savić
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000, Belgrade, Serbia.
| |
Collapse
|
25
|
Lei Y, Wang J, Wang D, Li C, Liu B, Fang X, You J, Guo M, Lu XY. SIRT1 in forebrain excitatory neurons produces sexually dimorphic effects on depression-related behaviors and modulates neuronal excitability and synaptic transmission in the medial prefrontal cortex. Mol Psychiatry 2020; 25:1094-1111. [PMID: 30705425 PMCID: PMC7192847 DOI: 10.1038/s41380-019-0352-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/08/2018] [Accepted: 12/20/2018] [Indexed: 12/20/2022]
Abstract
Sirtuin 1 (SIRT1), an NAD+-dependent deacetylase, is a key regulator of cellular metabolism. Recent genome-wide association studies identified genetic variants of SIRT1 linked to major depressive disorders. SIRT1 is widely expressed in the brain; however, neuronal substrates that mediate SIRT1 action on depressive behaviors remain largely unknown. Here we show that selective deletion of SIRT1 in forebrain excitatory neurons causes depression-like phenotypes in male but not female mice. AAV-Cre-mediated SIRT1 knockdown in the medial prefrontal cortex (mPFC) of adult male mice induces depressive-like behaviors. Whole-cell patch-clamp recordings demonstrate that loss of SIRT1 decreases intrinsic excitability and spontaneous excitatory synaptic transmission in layer V pyramidal neurons in the prelimbic mPFC. Consistent with neuronal hypoexcitability, SIRT1 knockout reduces mitochondrial density and expression levels of genes involved in mitochondrial biogenesis and dynamics in the prelimbic mPFC. When a SIRT1 activator (SRT2104) is injected into the mPFC or lateral ventricle of wild-type mice, it reverses chronic unpredictable stress-induced anhedonia and behavioral despair, indicating an antidepressant-like effect. These results suggest that SIRT1 in mPFC excitatory neurons is required for normal neuronal excitability and synaptic transmission and regulates depression-related behaviors in a sex-specific manner.
Collapse
Affiliation(s)
- Yun Lei
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jiangong Wang
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Dan Wang
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chen Li
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Bin Liu
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xing Fang
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jingjing You
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ming Guo
- Institute for Metabolic & Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xin-Yun Lu
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
- Department of Pharmacology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
26
|
Raith H, Schuelert N, Duveau V, Roucard C, Plano A, Dorner-Ciossek C, Ferger B. Differential effects of traxoprodil and S-ketamine on quantitative EEG and auditory event-related potentials as translational biomarkers in preclinical trials in rats and mice. Neuropharmacology 2020; 171:108072. [PMID: 32243874 DOI: 10.1016/j.neuropharm.2020.108072] [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: 10/10/2019] [Revised: 02/14/2020] [Accepted: 03/25/2020] [Indexed: 12/16/2022]
Abstract
Quantitative Electroencephalography (qEEG) and event-related potential (ERP) assessment have emerged as powerful tools to unravel translational biomarkers in preclinical and clinical psychiatric drug discovery trials. The aim of the present study was to compare the GluN2B negative allosteric modulator (NAM) traxoprodil (CP-101,606) with the unselective NMDA receptor channel blocker S-ketamine to give insight into central target engagement and differentiation on multiple EEG readouts. For qEEG recordings telemetric transmitters were implanted in male Wistar rats. Recorded EEG data were analyzed using fast Fourier transformation to determine power spectra and vigilance states. Additionally, body temperature and locomotor activity were assessed via telemetry. For recordings of auditory event-related potentials (AERP) male C57Bl/6J mice were chronically implanted with deep electrodes using a tethered system. Power spectral analysis revealed a significant increase in gamma power following ketamine treatment, whereas traxoprodil (6&18 mg/kg) induced an overall decrease primarily within alpha and beta bands. Additionally, ketamine disrupted sleep and enhanced time spent in wake vigilance states, whereas traxoprodil did not alter sleep-wake architecture. AERP and mismatch negativity (MMN) revealed that ketamine (10 mg/kg) selectively disrupts auditory deviance detection, whereas traxoprodil (6 mg/kg) did not alter MMN at clinically relevant doses. In contrast to ketamine treatment, traxoprodil did not produce hyperactivity and hypothermia. In conclusion, ketamine and traxoprodil showed very different effects on diverse EEG readouts differentiating selective GluN2B antagonism from non-selective pan-NMDA-R antagonists like ketamine. These readouts are thus perfectly suited to support drug discovery efforts on NMDA-R and understanding the different functions of NMDA-R subtypes.
Collapse
Affiliation(s)
- Henrike Raith
- Boehringer Ingelheim Pharma GmbH & Co. KG, CNS Diseases Research Germany, Birkendorferstr. 65, 88397, Biberach an der Riß, Germany.
| | - Niklas Schuelert
- Boehringer Ingelheim Pharma GmbH & Co. KG, CNS Diseases Research Germany, Birkendorferstr. 65, 88397, Biberach an der Riß, Germany.
| | - Venceslas Duveau
- SynapCell SAS, Biopolis and Institut Jean Roget, Université Joseph Fourier-Grenoble 1, Domaine de la merci, 38700, La Tronche, France.
| | - Corinne Roucard
- SynapCell SAS, Biopolis and Institut Jean Roget, Université Joseph Fourier-Grenoble 1, Domaine de la merci, 38700, La Tronche, France.
| | - Andrea Plano
- Plano Consulting, Georg-Schinbain-Str. 70, 88400, Biberach an der Riß, Germany.
| | - Cornelia Dorner-Ciossek
- Boehringer Ingelheim Pharma GmbH & Co. KG, CNS Diseases Research Germany, Birkendorferstr. 65, 88397, Biberach an der Riß, Germany.
| | - Boris Ferger
- Boehringer Ingelheim Pharma GmbH & Co. KG, CNS Diseases Research Germany, Birkendorferstr. 65, 88397, Biberach an der Riß, Germany.
| |
Collapse
|
27
|
Liu GL, Cui YF, Lu C, Zhao P. Ketamine a dissociative anesthetic: Neurobiology and biomolecular exploration in depression. Chem Biol Interact 2020; 319:109006. [DOI: 10.1016/j.cbi.2020.109006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/07/2020] [Accepted: 02/17/2020] [Indexed: 10/25/2022]
|
28
|
Mills RH, Wozniak JM, Vrbanac A, Campeau A, Chassaing B, Gewirtz A, Knight R, Gonzalez DJ. Organ-level protein networks as a reference for the host effects of the microbiome. Genome Res 2020; 30:276-286. [PMID: 31992612 PMCID: PMC7050531 DOI: 10.1101/gr.256875.119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
Connections between the microbiome and health are rapidly emerging in a wide range of diseases. However, a detailed mechanistic understanding of how different microbial communities are influencing their hosts is often lacking. One method researchers have used to understand these effects are germ-free (GF) mouse models. Differences found within the organ systems of these model organisms may highlight generalizable mechanisms that microbiome dysbioses have throughout the host. Here, we applied multiplexed, quantitative proteomics on the brains, spleens, hearts, small intestines, and colons of conventionally raised and GF mice, identifying associations to colonization state in over 7000 proteins. Highly ranked associations were constructed into protein-protein interaction networks and visualized onto an interactive 3D mouse model for user-guided exploration. These results act as a resource for microbiome researchers hoping to identify host effects of microbiome colonization on a given organ of interest. Our results include validation of previously reported effects in xenobiotic metabolism, the innate immune system, and glutamate-associated proteins while simultaneously providing organism-wide context. We highlight organism-wide differences in mitochondrial proteins including consistent increases in NNT, a mitochondrial protein with essential roles in influencing levels of NADH and NADPH, in all analyzed organs of conventional mice. Our networks also reveal new associations for further exploration, including protease responses in the spleen, high-density lipoproteins in the heart, and glutamatergic signaling in the brain. In total, our study provides a resource for microbiome researchers through detailed tables and visualization of the protein-level effects of microbial colonization on several organ systems.
Collapse
Affiliation(s)
- Robert H Mills
- Department of Pharmacology, University of California, San Diego, California 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California 92093, USA
- Department of Pediatrics, and Department of Computer Science and Engineering, University of California, San Diego, California 92093, USA
- Center for Microbiome Innovation, University of California, San Diego, California 92093, USA
| | - Jacob M Wozniak
- Department of Pharmacology, University of California, San Diego, California 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California 92093, USA
| | - Alison Vrbanac
- Department of Pediatrics, and Department of Computer Science and Engineering, University of California, San Diego, California 92093, USA
| | - Anaamika Campeau
- Department of Pharmacology, University of California, San Diego, California 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California 92093, USA
| | - Benoit Chassaing
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30303, USA
- Neuroscience Institute, Georgia State University, Atlanta, Georgia 30303, USA
- INSERM, U1016, 75014 Paris, France
- Université de Paris, 75006 Paris, France
| | - Andrew Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia 30303, USA
| | - Rob Knight
- Department of Pediatrics, and Department of Computer Science and Engineering, University of California, San Diego, California 92093, USA
- Center for Microbiome Innovation, University of California, San Diego, California 92093, USA
| | - David J Gonzalez
- Department of Pharmacology, University of California, San Diego, California 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, California 92093, USA
- Center for Microbiome Innovation, University of California, San Diego, California 92093, USA
| |
Collapse
|
29
|
Yao R, Wang H, Yuan M, Wang G, Wu C. Efficacy and safety of riluzole for depressive disorder: A systematic review and meta-analysis of randomized placebo-controlled trials. Psychiatry Res 2020; 284:112750. [PMID: 31958711 DOI: 10.1016/j.psychres.2020.112750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/26/2019] [Accepted: 01/01/2020] [Indexed: 02/08/2023]
Abstract
Glutamatergic modulators may have therapeutic potential in the treatment of depressive disorder (DD), riluzole, as a modulating drug of the glutamatergic system, its antidepressant efficacy and safety of riluzole for DD are inconsistent. This meta-analysis was performed to determine the efficacy and safety of riluzole used for DD. A systematic literature search was performed using PubMed, Embase, Cochrane Library, Web of Science, VIP and other databases from 1980 to 2019. The primary outcome was change in depression severity and meta-analysis was performed using comprehensive meta-analysis software. Seven randomized controlled trials (RCTs) were included. There was some difference in depression severity change in riluzole-citalopram therapy. No significant differences were observed in response rate, remission rate, relapse rate and adverse events, while, the relapse time in riluzole group was longer than placebo group. In this meta-analysis riluzole showed no antidepressant efficacy compared to placebo in monotherapy or riluzole-ketamine combined therapy, while it might relieve depression severity to some extent in riluzole-citalopram therapy. Furthermore, riluzole showed favorable safety for DD. The longer relapse time of riluzole group might have clinical significance to some extent, although this had no statistical difference. More studies are needed to clarify the potential association between riluzole and DD.
Collapse
Affiliation(s)
- Ruzhan Yao
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane Chengdu, Sichuan 610041, China
| | - Haiquan Wang
- Department of Neurosurgery, The People's Hospital of Guang'an City, Guangan 638001, China
| | - Mingqi Yuan
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guanglin Wang
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane Chengdu, Sichuan 610041, China.
| | - Chengxi Wu
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane Chengdu, Sichuan 610041, China
| |
Collapse
|
30
|
Camp AA, Yun J, Chambers SA, Haeba MH, LeBlanc GA. Involvement of glutamate and serotonin transmitter systems in male sex determination in Daphnia pulex. JOURNAL OF INSECT PHYSIOLOGY 2020; 121:104015. [PMID: 31930975 PMCID: PMC7098118 DOI: 10.1016/j.jinsphys.2020.104015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 05/30/2023]
Abstract
Environmental sex determination occurs in many organisms, however the means by which environmental stimuli are translated into endocrine messages remains poorly understood. The N-methyl-ᴅ-aspartate receptor (NMDAR) was evaluated as a candidate neural sensor of environmental signals linking environmental cues to endocrine responses using the crustacean Daphnia pulex. NMDAR agonists, modulators, and antagonists were evaluated for their ability to impact D. pulex male sex determination during early stages of reproductive maturity under conditions that simulated seasonal change. The antagonists MK-801 and desipramine significantly increased male sex determination. Both chemicals are also modulators of serotonergic and noradrenergic systems, thus, we evaluated several modulators of monoamine neurotransmission in an effort to discern which signaling pathways might contribute to male sex determination. Compounds that altered serotonergic signaling also stimulated male sex determination. The involvement of the glutamate and monoamine signaling in male sex determination was supported by the increase in mRNA levels of related receptors and transporters under conditions that stimulate male sex determination. Further, mRNA levels of components of the terminal endocrine pathway responsible for male sex determination were also elevated under stimulatory conditions. Overall, we provide evidence that glutamatergic and serotonergic systems function upstream of the endocrine regulation of male sex determination in early life stage daphnids.
Collapse
Affiliation(s)
- Allison A Camp
- North Carolina State University, Department of Biological Sciences, Raleigh, NC 27695, USA
| | - Jeonga Yun
- North Carolina State University, Department of Biological Sciences, Raleigh, NC 27695, USA
| | - Samantha A Chambers
- North Carolina State University, Department of Biological Sciences, Raleigh, NC 27695, USA
| | - Maher H Haeba
- North Carolina State University, Department of Biological Sciences, Raleigh, NC 27695, USA
| | - Gerald A LeBlanc
- North Carolina State University, Department of Biological Sciences, Raleigh, NC 27695, USA.
| |
Collapse
|
31
|
Poletti S, Mazza MG, Vai B, Lorenzi C, Colombo C, Benedetti F. Proinflammatory Cytokines Predict Brain Metabolite Concentrations in the Anterior Cingulate Cortex of Patients With Bipolar Disorder. Front Psychiatry 2020; 11:590095. [PMID: 33363485 PMCID: PMC7753118 DOI: 10.3389/fpsyt.2020.590095] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Bipolar disorder (BD) is a severe psychiatric illness characterized by abnormalities in the immune/inflammatory function and in brain metabolism. Evidences suggest that inflammation may affect the levels of brain metabolites as measured by single-proton magnetic resonance spectroscopy (1H-MRS). The aim of the study was to investigate whether a wide panel of inflammatory markers (i.e., cytokines, chemokines, and growth factors) can predict brain metabolite concentrations of glutamate, myo-inositol, N-acetylaspartate, and glutathione in a sample of 63 bipolar patients and 49 healthy controls. Three cytokines influenced brain metabolite concentrations: IL-9 positively predicts glutamate, IL-1β positively predicts Myo-inositol, and CCL5 positively predicts N-acetylaspartate concentrations. Furthermore, patients showed higher concentrations of glutamate, Myo-inositol, and glutathione and lower concentrations of N-acetylaspartate in respect to healthy controls. Our results confirm that inflammation in BD alters brain metabolism, through mechanisms possibly including the production of reactive oxygen species and glia activation.
Collapse
Affiliation(s)
- Sara Poletti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Mario Gennaro Mazza
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Benedetta Vai
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Cristina Lorenzi
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Colombo
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Benedetti
- Psychiatry & Clinical Psychobiology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
32
|
Gruenbaum BF, Kutz R, Zlotnik A, Boyko M. Blood glutamate scavenging as a novel glutamate-based therapeutic approach for post-stroke depression. Ther Adv Psychopharmacol 2020; 10:2045125320903951. [PMID: 32110376 PMCID: PMC7026819 DOI: 10.1177/2045125320903951] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 12/31/2019] [Indexed: 12/16/2022] Open
Abstract
Post-stroke depression (PSD) is a major complication of stroke that significantly impacts functional recovery and quality of life. While the exact mechanism of PSD is unknown, recent attention has focused on the association of the glutamatergic system in its etiology and treatment. Minimizing secondary brain damage and neuropsychiatric consequences associated with excess glutamate concentrations is a vital part of stroke management. The blood glutamate scavengers, oxaloacetate and pyruvate, degrade glutamate in the blood to its inactive metabolite, 2-ketoglutarate, by the coenzymes glutamate-oxaloacetate transaminase (GOT) and glutamate-pyruvate transaminase (GPT), respectively. This reduction in blood glutamate concentrations leads to a subsequent shift of glutamate down its concentration gradient from the blood to the brain, thereby decreasing brain glutamate levels. Although there are not yet any human trials that support blood glutamate scavengers for clinical use, there is increasing evidence from animal research of their efficacy as a promising new therapeutic approach for PSD. In this review, we present recent evidence in the literature of the potential therapeutic benefits of blood glutamate scavengers for reducing PSD and other related neuropsychiatric conditions. The evidence reviewed here should be useful in guiding future clinical trials.
Collapse
Affiliation(s)
- Benjamin F Gruenbaum
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ruslan Kutz
- Division of Anesthesiology and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alexander Zlotnik
- Division of Anesthesiology and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Matthew Boyko
- Division of Anesthesiology and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| |
Collapse
|
33
|
Shaffer JJ, Mani M, Schmitz SL, Xu J, Owusu N, Wu D, Magnotta VA, Wemmie JA. Proton Exchange Magnetic Resonance Imaging: Current and Future Applications in Psychiatric Research. Front Psychiatry 2020; 11:532606. [PMID: 33192650 PMCID: PMC7542226 DOI: 10.3389/fpsyt.2020.532606] [Citation(s) in RCA: 4] [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: 02/04/2020] [Accepted: 09/02/2020] [Indexed: 12/20/2022] Open
Abstract
Proton exchange provides a powerful contrast mechanism for magnetic resonance imaging (MRI). MRI techniques sensitive to proton exchange provide new opportunities to map, with high spatial and temporal resolution, compounds important for brain metabolism and function. Two such techniques, chemical exchange saturation transfer (CEST) and T1 relaxation in the rotating frame (T1ρ), are emerging as promising tools in the study of neurological and psychiatric illnesses to study brain metabolism. This review describes proton exchange for non-experts, highlights the current status of proton-exchange MRI, and presents advantages and drawbacks of these techniques compared to more traditional methods of imaging brain metabolism, including positron emission tomography (PET) and MR spectroscopy (MRS). Finally, this review highlights new frontiers for the use of CEST and T1ρ in brain research.
Collapse
Affiliation(s)
- Joseph J Shaffer
- Department of Radiology, University of Iowa, Iowa City, IA, United States
| | - Merry Mani
- Department of Radiology, University of Iowa, Iowa City, IA, United States
| | - Samantha L Schmitz
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States
| | - Jia Xu
- Department of Radiology, University of Iowa, Iowa City, IA, United States
| | - Nana Owusu
- Department of Radiology, University of Iowa, Iowa City, IA, United States.,Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, United States.,Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - Dee Wu
- Department of Radiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Vincent A Magnotta
- Department of Radiology, University of Iowa, Iowa City, IA, United States.,Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Department of Biomedical Engineering, University of Iowa, Iowa City, IA, United States
| | - John A Wemmie
- Department of Psychiatry, University of Iowa, Iowa City, IA, United States.,Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA, United States.,Department of Veterans Affairs Medical Center, Iowa City, IA, United States.,Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, United States.,Department of Neurosurgery, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
34
|
Functional magnetic resonance spectroscopy in patients with schizophrenia and bipolar affective disorder: Glutamate dynamics in the anterior cingulate cortex during a working memory task. Eur Neuropsychopharmacol 2019; 29:222-234. [PMID: 30558824 DOI: 10.1016/j.euroneuro.2018.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 11/25/2018] [Accepted: 12/01/2018] [Indexed: 12/16/2022]
Abstract
The glutamate system is implicated in the pathophysiology of schizophrenia and mood disorders. Using functional magnetic resonance spectroscopy (1H-fMRS), it is possible to monitor glutamate dynamically in activated brain areas and may give a closer estimate of glutamatergic neurotransmission than standard magnetic resonance spectroscopy. 14 patients with schizophrenia, 15 patients with bipolar disorder II (BPII) and 14 healthy volunteers underwent a 15 min N-back task in a 48s block design during 1H-fMRS acquisition. Data from the first, second and third 16s group of 8 spectra for each block were analysed to measure levels of glutamate and Glx (glutamate + glutamine), scaled to total creatine (TCr), across averaged 0-back and 2-back conditions. A 6 × 3 repeated-measures analysis of variance (rmANOVA) demonstrated a significant main effect of time for Glx/TCr (P = 0.022). There was a significant increase in Glu/TCr (P = 0.004) and Glx/TCr (P < 0.001) between the final spectra of the 0-back and first spectra of the 2-back condition in the healthy control group only. 2 × 2 rmANOVA revealed a significant time by group interaction for Glx/TCr (P = 0.019) across the 0-back condition, with levels reducing in healthy controls and increasing in the schizophrenia group. While healthy volunteers showed significant increases in glutamatergic measures between task conditions, the lack of such a response in patients with schizophrenia and BPII may reflect deficits in glutamatergic neurotransmission. Abnormal increases during periods of relatively low executive load, without the same dynamic modulation as healthy volunteers with increasing task difficulty, further suggests underlying abnormalities of glutamatergic neurotransmission in schizophrenia.
Collapse
|
35
|
Matraszek-Gawron R, Chwil M, Terlecka P, Skoczylas MM. Recent Studies on Anti-Depressant Bioactive Substances in Selected Species from the Genera Hemerocallis and Gladiolus: A Systematic Review. Pharmaceuticals (Basel) 2019; 12:ph12040172. [PMID: 31775329 PMCID: PMC6958339 DOI: 10.3390/ph12040172] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/04/2019] [Accepted: 11/22/2019] [Indexed: 12/14/2022] Open
Abstract
Herbal therapy is a potential alternative applied to pharmacological alleviation of depression symptoms and treatment of this disorder, which is predicted by the World Health Organization (WHO) to be the most serious health problem worldwide over the next several years. It has been well documented that many herbs with psychotropic effects have far fewer side effects than a variety of pharmaceutical agents used by psychiatrists for the treatment of depression. This systematic review presents literature data on the antidepressant activity of representatives of the genera Hemerocallis (H. fulva and H. citrina Baroni, family Xanthorrhoeaceae) and Gladiolus (G. dalenii, family Iridaceae) and on biologically active compounds and their mechanisms of action to consider the application of herbal preparations supporting the treatment of depression.
Collapse
Affiliation(s)
- Renata Matraszek-Gawron
- Department of Botany and Plant Physiology, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland;
| | - Mirosława Chwil
- Department of Botany and Plant Physiology, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland;
- Correspondence: ; Tel.: +48-81-445-66-24
| | - Paulina Terlecka
- Chair and Department of Pneumology, Oncology and Allergology, Medical University of Lublin, 8 Jaczewskiego Street, 20-090 Lublin, Poland;
| | - Michał M. Skoczylas
- Department of Diagnostic Imaging and Interventional Radiology, Pomeranian Medical University in Szczecin, 1 Unii Lubelskiej Street, 71-252 Szczecin, Poland;
| |
Collapse
|
36
|
Dehghani M, Do KQ, Magistretti P, Xin L. Lactate measurement by neurochemical profiling in the dorsolateral prefrontal cortex at 7T: accuracy, precision, and relaxation times. Magn Reson Med 2019; 83:1895-1908. [PMID: 31729080 DOI: 10.1002/mrm.28066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/17/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE This assesses the potential of measuring lactate in the human brain using three non-editing MRS methods at 7T and compares the accuracy and precision of the methods. METHODS 1 H MRS data were measured in the right dorsolateral prefrontal cortex using a semi-adiabatic spin-echo full-intensity acquired localized sequence with three different protocols: (I) TE = 16 ms, (II) TE = 110 ms, and (III) TE = 16 ms, TI = 300 ms. T1 and T2 relaxation times of lactate were also measured. Simulated spectra were generated for three protocols with known concentrations, using a range of spectral linewidths and SNRs to assess the effect of data quality on the measurement precision and accuracy. RESULTS Lactate was quantified in all three protocols with mean Cramér-Rao lower bound of 8% (I), 13% (II), and 7% (III). The T1 and T2 relaxation times of lactate were 1.9 ± 0.2 s and 94 ± 13 ms, respectively. Simulations predicted a spectral linewidth-associated underestimation of lactate measurement. Simulations, phantom and in vivo results showed that protocol II was most affected by this underestimation. In addition, the estimation error was insensitive to a broad range of spectral linewidth with protocol I. Within-session coefficient of variances of lactate were 6.1 ± 7.9% (I), 22.3 ± 12.3% (II), and 5.1 ± 5.4% (III), respectively. CONCLUSION We conclude that protocols I and III have the potential to measure lactate at 7T with good reproducibility, whereas the measurement accuracy and precision depend on spectral linewidth and SNR, respectively. Moreover, simulation is valuable for the optimization of measurement protocols in future study design and the correction for measurement bias.
Collapse
Affiliation(s)
- Masoumeh Dehghani
- Center for Psychiatric Neuroscience (CNP), Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience (CNP), Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland
| | - Pierre Magistretti
- Center for Psychiatric Neuroscience (CNP), Department of Psychiatry, Lausanne University Hospital-CHUV, Prilly-Lausanne, Switzerland.,BESE Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia.,Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Lijing Xin
- Animal Imaging and Technology Core (AIT), Center for Biomedical Imaging (CIBM), Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
37
|
Dextromethorphan/Bupropion: A Novel Oral NMDA (N-methyl-d-aspartate) Receptor Antagonist with Multimodal Activity. CNS Spectr 2019; 24:461-466. [PMID: 31566163 DOI: 10.1017/s1092852919001470] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Although currently available antidepressants increase monoamine levels soon after the start of treatment, therapeutic benefits are often delayed by several weeks and the majority of patients with major depressive disorder fail to achieve an adequate response to first- or second-line therapies targeting monoamines. The recent approval of the NMDA (N-methyl-d-aspartate) antagonist esketamine given intranasally for treatment-resistant depression has reinforced the need for agents with rapid onset with alternate mechanisms of action. Dextromethorphan/bupropion, an investigational medicine currently in development, is one such candidate.
Collapse
|
38
|
The Effect of Glutamatergic Modulators on Extracellular Glutamate: How Does this Information Contribute to the Discovery of Novel Antidepressants? Curr Ther Res Clin Exp 2019; 91:25-32. [PMID: 31871505 PMCID: PMC6911922 DOI: 10.1016/j.curtheres.2019.100566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 08/29/2019] [Indexed: 01/19/2023] Open
Abstract
The complexity of glutamatergic signaling challenges glutamate modulator usage. Functional biomarkers are needed to understand the MOA of glutamate modulators. Evaluating drug effect on EAATs' kinetics may add to antidepressant discovery.
Background In the search for new antidepressants, clinical researchers have been using drugs that simultaneously modulate multiple targets. During preclinical and clinical trials, the glutamatergic modulators riluzole and ketamine have received particular attention. Glutamatergic agents have a modulatory effect on synaptic transmission, so they can act on both neurons and astrocytes. In addition to influencing the quantity of glutamate released, these modulators can also affect the expression, localization, and functionality of glutamate-binding sites. Objective This review discusses the complexity of the glutamatergic system, the ambiguity of data regarding glutamate levels in patients with depression, as well as the mechanisms of action for riluzole and ketamine, which includes their relation to the physiology of glutamatergic transmission. The principal aim is to contribute to the development of novel glutamatergic antidepressant medications whilst emphasizing the need for innovative approaches that evaluate their effects on extracellular glutamate. Methods Literature was obtained via PubMed by searching the term depression in combination with each of the following terms: riluzole, ketamine, and glutamate. The search was restricted to full-text articles published in English between 1985 and 2018 relating to both the modulatory mechanisms of glutamatergic-binding proteins and the antidepressant actions of these medicines. Articles about mechanisms associated with synaptic plasticity and antidepressant effects were excluded. Results Although experimental data relates glutamatergic signaling to the pathophysiology of major depression and bipolar disorder, the role of glutamate—as well as its extracellular concentration in patients with said disorders—is still unclear. Riluzole's antidepressant action is ascribed to its capacity to reduce glutamate levels in the synaptic cleft, and ketamine's effect has been associated with increased extracellular glutamate levels. Conclusions The strategy of using glutamatergic modulators as therapeutic agents requires a better understanding of the role of glutamate in the pathophysiology of depression. Gaining such understanding is a challenge because it entails evaluating different targets as well as the effects of these modulators on the kinetics of glutamate uptake. Essentially, glutamate transport is a dynamic process and, currently, it is still necessary to develop new approaches to assay glutamate in the synaptic cleft. ORCID: 0000-0002-3358-6939.
Collapse
|
39
|
Pham TH, Gardier AM. Fast-acting antidepressant activity of ketamine: highlights on brain serotonin, glutamate, and GABA neurotransmission in preclinical studies. Pharmacol Ther 2019; 199:58-90. [DOI: 10.1016/j.pharmthera.2019.02.017] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/25/2019] [Indexed: 12/13/2022]
|
40
|
Humo M, Lu H, Yalcin I. The molecular neurobiology of chronic pain-induced depression. Cell Tissue Res 2019; 377:21-43. [PMID: 30778732 DOI: 10.1007/s00441-019-03003-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 02/01/2019] [Indexed: 12/18/2022]
Abstract
The increasing number of individuals with comorbidities poses an urgent need to improve the management of patients with multiple co-existing diseases. Among these comorbidities, chronic pain and mood disorders, two long-lasting disabling conditions that significantly reduce the quality of life, could be cited first. The recent development of animal models accelerated the studies focusing on the underlying mechanisms of the chronic pain and depression/anxiety comorbidity. This review provides an overview of clinical and pre-clinical studies performed over the past two decades addressing the molecular aspects of the comorbid relationship of chronic pain and depression. We thus focused on the studies that investigated the molecular characteristics of the comorbid relationship between chronic pain and mood disorders, especially major depressive disorders, from the genetic and epigenetic point of view to key neuromodulators which have been shown to play an important role in this comorbidity.
Collapse
Affiliation(s)
- Muris Humo
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique et Université de Strasbourg, 67000, Strasbourg, France
| | - Han Lu
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique et Université de Strasbourg, 67000, Strasbourg, France.,Faculty of Biology and Bernstein Center Freiburg, University of Freiburg, D-79104, Freiburg, Germany
| | - Ipek Yalcin
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique et Université de Strasbourg, 67000, Strasbourg, France.
| |
Collapse
|
41
|
Riva-Posse P, Reiff CM, Edwards JA, Job GP, Galendez GC, Garlow SJ, Saah TC, Dunlop BW, McDonald WM. Blood pressure safety of subanesthetic ketamine for depression: A report on 684 infusions. J Affect Disord 2018. [PMID: 29525051 DOI: 10.1016/j.jad.2018.02.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The dissociative anesthetic agent ketamine is increasingly being utilized to treat depression, despite not having FDA (Food and Drug Administration) approval for this indication. There are many questions about the potential risks of this treatment and hence the proper setting and degree of monitoring required to ensure patient safety. There is limited data about the cardiovascular safety of ketamine when administered at subanesthetic doses to treat depression. METHODS 66 patients in the Department of Psychiatry at Emory University received a total of 684 ketamine infusions between 2014 and 2016. Ketamine was dosed at 0.5 mg/kg body weight and infused over 40 min. Blood pressure was measured every 10 min during the infusions and every 15 min thereafter. RESULTS Mean age of the patients was 56.7 years, 87.9% had unipolar depression and 36.1% had essential hypertension. No infusions were discontinued due to instability of vital signs, adverse physiological consequences or acute psychotomimetic effects. The biggest increases in blood pressure were measured at 30 min (systolic 3.28 mmHg, diastolic 3.17 mmHg). Hypertensive patients had higher blood pressure peaks during the infusions. Blood pressures returned to baseline during post-infusion monitoring. There was no development of tolerance to the blood pressure elevating effects of ketamine between the first and sixth infusions. LIMITATIONS This is a single site, retrospective analysis, of patients who were spontaneously seeking clinical care. CONCLUSIONS The blood pressure changes observed when ketamine is administered over 40 min at 0.5 mg/kg for the treatment of depression are small, well tolerated and clinically insignificant.
Collapse
Affiliation(s)
- Patricio Riva-Posse
- Emory University Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA.
| | - Collin M Reiff
- Emory University Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA
| | - Johnathan A Edwards
- Emory University Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA
| | - Gregory P Job
- Emory University Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA
| | - Gail C Galendez
- Emory University Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA
| | - Steven J Garlow
- University of Wisconsin Department of Psychiatry, Madison, WI, USA
| | - Tammy C Saah
- Integrated Clinical Neurosciences and Silicon Valley TMS, San Jose, CA, USA
| | - Boadie W Dunlop
- Emory University Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA
| | - William M McDonald
- Emory University Department of Psychiatry and Behavioral Sciences, Atlanta, GA, USA
| |
Collapse
|
42
|
Porcelli S, Marsano A, Caletti E, Sala M, Abbiati V, Bellani M, Perlini C, Rossetti MG, Mandolini GM, Pigoni A, Paoli RA, Piccin S, Lazzaretti M, Fabbro D, Damante G, Bonivento C, Ferrari C, Rossi R, Pedrini L, Serretti A, Brambilla P. Temperament and Character Inventory in Bipolar Disorder versus Healthy Controls and Modulatory Effects of 3 Key Functional Gene Variants. Neuropsychobiology 2018; 76:209-221. [PMID: 30041166 DOI: 10.1159/000490955] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/18/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Bipolar disorder (BD) has been associated with temperamental and personality traits, although the relationship is still to be fully elucidated. Several studies investigated the genetic basis of temperament and character, identifying catechol-O-methyltransferase (COMT), brain derived neurotrophic factor (BDNF), and serotonin transporter (5-HTT) gene variants as strong candidates. METHODS In the GECO-BIP study, 125 BD patients and 173 HC were recruited. Subjects underwent to a detailed assessment and the temperament and character inventory 125 items (TCI) was administrated. Three functional genetic variants within key candidate genes (COMT rs4680, BDNF rs6265, and the serotonin-transporter-linked polymorphic region (5-HTTLPR)) were genotyped. Univariate and multivariate analyses were performed. RESULTS Compared to HC, BD patients showed higher scores in novelty seeking (NS; p = 0.001), harm avoidance (HA; p < 0.001), and self transcendence (St; p < 0.001), and lower scores in self directness (p < 0.001) and cooperativeness (p < 0.001) TCI dimensions. Concerning the genetic analyses, COMT rs4680 was associated with NS in the total sample (p = 0.007) and in the male subsample (p = 0.022). When performing the analysis in the HC and BD samples, the association was confirmed only in HC (p = 0.012), and in the HC male subgroup in particular (p = 0.004). BDNF rs6265 was associated with St in the BD group (p = 0.017). CONCLUSION COMT rs4680 may modulate NS in males in the general population. This effect was not detected in BD patients, probably because BD alters the neurobiological basis of some TCI dimensions. BDNF rs6265 seems to modulate St TCI dimension only in BD patients, possibly modulating the previously reported association between rs6265 and BD treatment response. Further studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Stefano Porcelli
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Agnese Marsano
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Elisabetta Caletti
- Department of Neurosciences and Mental Health, IRCCS Fondazione Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Michela Sala
- Department of Mental Health, Azienda Sanitaria Locale Alessandria, Alessandria, Italy
| | - Vera Abbiati
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Marcella Bellani
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry and Clinical Psychology, University of Verona, Verona, Italy.,UOC Psychiatry, Azienda Ospedaliera Universitaria Integrata Verona (AOUI), Verona, Italy
| | - Cinzia Perlini
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry and Clinical Psychology, University of Verona, Verona, Italy.,UOC Psychiatry, Azienda Ospedaliera Universitaria Integrata Verona (AOUI), Verona, Italy
| | - Maria Gloria Rossetti
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Psychiatry and Clinical Psychology, University of Verona, Verona, Italy
| | - Gian Mario Mandolini
- Department of Neurosciences and Mental Health, IRCCS Fondazione Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Alessandro Pigoni
- Department of Neurosciences and Mental Health, IRCCS Fondazione Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Riccardo Augusto Paoli
- Department of Neurosciences and Mental Health, IRCCS Fondazione Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Sara Piccin
- Scientific Institute IRCCS "Eugenio Medea," Polo FVG, San Vito al Tagliamento, Pordenone, Italy
| | - Matteo Lazzaretti
- Department of Neurosciences and Mental Health, IRCCS Fondazione Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Dora Fabbro
- Institute of Medical Genetics, Department of Laboratory Medicine, University of Udine, Udine, Italy
| | - Giuseppe Damante
- Institute of Medical Genetics, Department of Laboratory Medicine, University of Udine, Udine, Italy
| | - Carolina Bonivento
- Unit of Psychiatry, Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Clarissa Ferrari
- Service of Statistics, IRCCS Centro San Giovanni di Dio FBF, Brescia, Italy
| | - Roberta Rossi
- Unit of Psychiatry, IRCCS Centro San Giovanni di Dio FBF, Brescia, Italy
| | - Laura Pedrini
- Unit of Psychiatry, IRCCS Centro San Giovanni di Dio FBF, Brescia, Italy
| | - Alessandro Serretti
- Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy
| | - Paolo Brambilla
- Department of Neurosciences and Mental Health, IRCCS Fondazione Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy.,Department of Psychiatry and Behavioral Sciences, University of Texas at Houston, Houston, Texas, USA
| | | |
Collapse
|
43
|
Reed JL, Nugent AC, Furey ML, Szczepanik JE, Evans JW, Zarate CA. Ketamine normalizes brain activity during emotionally valenced attentional processing in depression. NEUROIMAGE-CLINICAL 2018; 20:92-101. [PMID: 30094160 PMCID: PMC6070691 DOI: 10.1016/j.nicl.2018.07.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/15/2018] [Accepted: 07/04/2018] [Indexed: 01/01/2023]
Abstract
Background An urgent need exists for faster-acting pharmacological treatments in major depressive disorder (MDD). The glutamatergic modulator ketamine has been shown to have rapid antidepressant effects, but much remains unknown about its mechanism of action. Functional MRI (fMRI) can be used to investigate how ketamine impacts brain activity during cognitive and emotional processing. Methods This double-blind, placebo-controlled, crossover study of 33 unmedicated participants with MDD and 26 healthy controls (HCs) examined how ketamine affected fMRI activation during an attentional bias dot probe task with emotional face stimuli across multiple time points. A whole brain analysis was conducted to find regions with differential activation associated with group, drug session, or dot probe task-specific factors (emotional valence and congruency of stimuli). Results A drug session by group interaction was observed in several brain regions, such that ketamine had opposite effects on brain activation in MDD versus HC participants. Additionally, there was a similar finding related to emotional valence (a drug session by group by emotion interaction) in a large cluster in the anterior cingulate and medial frontal cortex. Conclusions The findings show a pattern of brain activity in MDD participants following ketamine infusion that is similar to activity observed in HCs after placebo. This suggests that ketamine may act as an antidepressant by normalizing brain function during emotionally valenced attentional processing. Clinical trial NCT#00088699: https://www.clinicaltrials.gov/ct2/show/NCT00088699 The effects of ketamine versus placebo on brain activation were studied using fMRI. MDD and healthy participants were tested on an fMRI emotion-based attentional task. Ketamine had opposite effects on brain activity in MDD versus healthy participants. In MDD, brain activity post-ketamine was similar to healthy controls post-placebo. These findings suggest that ketamine may act by normalizing brain function.
Collapse
Affiliation(s)
- Jessica L Reed
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.
| | - Allison C Nugent
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.
| | - Maura L Furey
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States; Janssen Pharmaceuticals of Johnson and Johnson Inc., San Diego, CA, United States.
| | - Joanna E Szczepanik
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.
| | - Jennifer W Evans
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.
| | - Carlos A Zarate
- Experimental Therapeutics & Pathophysiology Branch, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
44
|
Cattaneo C, Müller T, Bonizzoni E, Lazzeri G, Kottakis I, Keywood C. Long-Term Effects of Safinamide on Mood Fluctuations in Parkinson's Disease. JOURNAL OF PARKINSONS DISEASE 2018; 7:629-634. [PMID: 28777756 PMCID: PMC5676861 DOI: 10.3233/jpd-171143] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background: Mood disorders are very frequent in Parkinson’s Disease (PD), and their effective treatment is still a major unresolved issue: growing evidence suggests that glutamatergic system dysfunction is directly involved. Safinamide is a drug with an innovative mechanism of action, dopaminergic and non-dopaminergic, that includes the reversible inhibition of the monoamine oxidase-B (MAO-B) enzyme and the modulation of excessive glutamate release through the use- and state-dependent blockade of the sodium channels. Objective: To investigate the effects of safinamide on mood over two-year treatment in PD patients with motor fluctuations. Methods: This was a post-hoc analysis of the data from studies 016 and 018. The analysis focused on outcomes related to mood, namely: scores of the “Emotional well-being” domain of the Parkinson’s Disease Questionnaire (PDQ-39), scores of the GRID Hamilton Rating Scale for Depression (GRID-HAMD) and the proportion of patients reporting depression as an adverse event over the entire treatment period. Results: Safinamide, compared to placebo, significantly improved the PDQ-39 “Emotional well-being” domain after6-months (p = 0.0067) and 2 years (p = 0.0006), as well as the GRID-HAMD (p = 0.0408 after 6 months and p = 0.0027 after 2 years). Significantly fewer patients in the safinamide group, compared to placebo, experienced depression as adverse event (p = 0.0444 after 6 months and p = 0.0057 after 2 years). Conclusion: The favorable effect of safinamide on mood may be explained by the improvement in wearing off and by its modulation of glutamatergic hyperactivity and reversible MAO-B inhibition. Prospective studies are warranted to investigate this potential benefit.
Collapse
Affiliation(s)
| | - Thomas Müller
- Department of Neurology, St. Joseph Hospital Berlin-Weißensee, Berlin, Germany
| | - Erminio Bonizzoni
- Department of Clinical Science and Community, Section of Medical Statistics and Biometry "GA Maccacaro", University of Milan, Milan, Italy
| | | | | | | |
Collapse
|
45
|
Effects of systemic glutamatergic manipulations on conditioned eyeblink responses and hyperarousal in a rabbit model of post-traumatic stress disorder. Behav Pharmacol 2018; 28:565-577. [PMID: 28799954 DOI: 10.1097/fbp.0000000000000333] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Glutamatergic dysfunction is implicated in many neuropsychiatric conditions, including post-traumatic stress disorder (PTSD). Glutamate antagonists have shown some utility in treating PTSD symptoms, whereas glutamate agonists may facilitate cognitive behavioral therapy outcomes. We have developed an animal model of PTSD, based on conditioning of the rabbit's eyeblink response, that addresses two key features: conditioned responses (CRs) to cues associated with an aversive event and a form of conditioned hyperarousal referred to as conditioning-specific reflex modification (CRM). The optimal treatment to reduce both CRs and CRM is unpaired extinction. The goals of the study were to examine whether treatment with the N-methyl-D-aspartate glutamate receptor antagonist ketamine could reduce CRs and CRM, and whether the N-methyl-D-aspartate agonist D-cycloserine combined with unpaired extinction treatment could enhance the extinction of both. Administration of a single dose of subanesthetic ketamine had no significant immediate or delayed effect on CRs or CRM. Combining D-cycloserine with a single day of unpaired extinction facilitated extinction of CRs in the short term while having no impact on CRM. These results caution that treatments may improve one aspect of the PTSD symptomology while having no significant effects on other symptoms, stressing the importance of a multiple-treatment approach to PTSD and of animal models that address multiple symptoms.
Collapse
|
46
|
Hlavacova N, Li Y, Pehrson A, Sanchez C, Bermudez I, Csanova A, Jezova D, Franklin M. Effects of vortioxetine on biomarkers associated with glutamatergic activity in an SSRI insensitive model of depression in female rats. Prog Neuropsychopharmacol Biol Psychiatry 2018; 82:332-338. [PMID: 29269186 DOI: 10.1016/j.pnpbp.2017.07.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/09/2017] [Accepted: 07/09/2017] [Indexed: 11/27/2022]
Abstract
The aim of this study was to investigate the antidepressant activity of vortioxetine in a tryptophan (TRP) depletion female rat model of depression and compare it to that of paroxetine using doses that fully occupy the serotonin transporter (SERT). We evaluated the effects of vortioxetine on potential biomarkers associated with TRP depletion including serum aldosterone, corticosterone and IL-6 levels together with indirect indicators of glutamate neurotransmission. Female Sprague-Dawley rats were randomized to control, low TRP, low TRP/paroxetine or low TRP/vortioxetine groups. Vortioxetine and paroxetine were administered via diet (10mg/kg/day) and drinking water (10mg/kg/day) respectively for 14days. Vortioxetine but not paroxetine reversed TRP depletion-induced depressive-like behavior. Vortioxetine reduced TRP depletion-induced increases of serum corticosterone, aldosterone, IL-6 and N-methyl-d-aspartate and α7-nicotinic acetylcholine receptor expression in the amygdala and hippocampus, respectively. Paroxetine demonstrated little effect except a reduction of aldosterone. Vortioxetine but not paroxetine reversed TRP depletion-induced reductions of serum and brain kynurenic acid. In conclusion, vortioxetine, but not paroxetine, enabled reversals of TRP depletion-induced changes of depression-like behavior and markers of glutamatergic activity. These observations support the hypothesis that vortioxetine's antidepressant activity may involve mechanisms beyond SERT inhibition.
Collapse
Affiliation(s)
- N Hlavacova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Y Li
- Department of Neuroscience, Lundbeck Research USA, Inc., Paramus, NJ, USA
| | - A Pehrson
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Inc., Paramus, NJ, USA
| | - C Sanchez
- Department of Neuroscience, Lundbeck Research USA, Inc., Paramus, NJ, USA
| | - I Bermudez
- School of Life Sciences, Oxford Brookes University, Oxford, UK
| | - A Csanova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - D Jezova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - M Franklin
- School of Life Sciences, Oxford Brookes University, Oxford, UK.
| |
Collapse
|
47
|
Engaging homeostatic plasticity to treat depression. Mol Psychiatry 2018; 23:26-35. [PMID: 29133952 DOI: 10.1038/mp.2017.225] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/11/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022]
Abstract
Major depressive disorder (MDD) is a complex and heterogeneous mood disorder, making it difficult to develop a generalized, pharmacological therapy that is effective for all who suffer from MDD. Through the fortuitous discovery of N-methyl-D-aspartate receptor (NMDAR) antagonists as effective antidepressants, we have gained key insights into how antidepressant effects can be produced at the circuit and molecular levels. NMDAR antagonists act as rapid-acting antidepressants such that relief from depressive symptoms occurs within hours of a single injection. The mode of action of NMDAR antagonists seemingly relies on their ability to activate protein-synthesis-dependent homeostatic mechanisms that restore top-down excitatory connections. Recent evidence suggests that NMDAR antagonists relieve depressive symptoms by forming new synapses resulting in increased excitatory drive. This event requires the mammalian target of rapamycin complex 1 (mTORC1), a signaling pathway that regulates synaptic protein synthesis. Herein, we review critical studies that shed light on the action of NMDAR antagonists as rapid-acting antidepressants and how they engage a neuron's or neural network's homeostatic mechanisms to self-correct. Recent studies notably demonstrate that a shift in γ-amino-butyric acid receptor B (GABABR) function, from inhibitory to excitatory, is required for mTORC1-dependent translation with NMDAR antagonists. Finally, we discuss how GABABR activation of mTORC1 helps resolve key discrepancies between rapid-acting antidepressants and local homeostatic mechanisms.
Collapse
|
48
|
Nasca C, Bigio B, Zelli D, de Angelis P, Lau T, Okamoto M, Soya H, Ni J, Brichta L, Greengard P, Neve RL, Lee FS, McEwen BS. Role of the Astroglial Glutamate Exchanger xCT in Ventral Hippocampus in Resilience to Stress. Neuron 2017; 96:402-413.e5. [PMID: 29024663 DOI: 10.1016/j.neuron.2017.09.020] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 08/10/2017] [Accepted: 09/13/2017] [Indexed: 12/23/2022]
Abstract
We demonstrate that stress differentially regulates glutamate homeostasis in the dorsal and ventral hippocampus and identify a role for the astroglial xCT in ventral dentate gyrus (vDG) in stress and antidepressant responses. We provide an RNA-seq roadmap for the stress-sensitive vDG. The transcription factor REST binds to xCT promoter in co-occupancy with the epigenetic marker H3K27ac to regulate expression of xCT, which is also reduced in a genetic mouse model of inherent susceptibility to depressive-like behavior. Pharmacologically, modulating histone acetylation with acetyl-L-carnitine (LAC) or acetyl-N-cysteine (NAC) rapidly increases xCT and activates a network with mGlu2 receptors to prime an enhanced glutamate homeostasis that promotes both pro-resilient and antidepressant-like responses. Pharmacological xCT blockage counteracts NAC prophylactic effects. GFAP+-Cre-dependent overexpression of xCT in vDG mimics pharmacological actions in promoting resilience. This work establishes a mechanism by which vDG protection leads to stress resilience and antidepressant responses via epigenetic programming of an xCT-mGlu2 network.
Collapse
Affiliation(s)
- Carla Nasca
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA.
| | - Benedetta Bigio
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Danielle Zelli
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Paolo de Angelis
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Timothy Lau
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA
| | - Masahiro Okamoto
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA; Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki 305-8574, Japan
| | - Hideyo Soya
- Department of Sports Neuroscience, Advanced Research Initiative for Human High Performance (ARIHHP), Faculty of Health and Sport Sciences, University of Tsukuba, Tennodai, Tsukuba, Ibaraki 305-8574, Japan; Laboratory of Exercise Biochemistry and Neuroendocrinology, Faculty of Health and Sport Sciences, University of Tsukuba, Ibaraki 305-8574, Japan
| | - Jason Ni
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Lars Brichta
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Paul Greengard
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY 10065, USA
| | - Rachael L Neve
- Gene Delivery Technology Core, Massachusetts General Hospital, 65 Landsdowne Street, Cambridge, MA 02139, USA
| | - Francis S Lee
- Sackler Institute for Developmental Psychobiology, Department of Psychiatry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
49
|
Jiang Y, Wang Y, Sun X, Lian B, Sun H, Wang G, Du Z, Li Q, Sun L. Short- and long-term antidepressant effects of ketamine in a rat chronic unpredictable stress model. Brain Behav 2017; 7:e00749. [PMID: 28828210 PMCID: PMC5561310 DOI: 10.1002/brb3.749] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE This research was aimed to evaluate the behaviors of short- or long-term antidepressant effects of ketamine in rats exposed to chronic unpredictable stress (CUS). BACKGROUND Ketamine, a glutamate noncompetitive NMDA receptor antagonist, regulates excitatory amino acid functions, such as anxiety disorders and major depression, and plays an important role in synaptic plasticity and learning and memory. METHODS After 42 days of CUS model, male rats received either a single injection of ketamine (10 mg/kg; day 43) or 15 daily injections (days 43-75). The influence of ketamine on behavioral reactivity was assessed 24 hr (short-term) or 7 weeks after ketamine treatment (long-term). Behavioral tests used to assess the effects of these treatments included the sucrose preference (SP), open field (OF), elevated plus maze (EPM), forced swimming (FS), and water maze (WM) to detect anxiety-like behavior (OF and EPM), forced swimming (FS), and water maze (WM). Results: Short-term ketamine administration resulted in increases of body weight gain, higher sensitivity to sucrose, augmented locomotor activity in the OF, more entries into the open arms of the EPM, along increased activity in the FS test; all responses indicative of reductions in depression/despair in anxiety-eliciting situations. No significant differences in these behaviors were obtained under conditions of long-term ketamine administration (p > .05). The CUS + Ketamine group showed significantly increased activity as compared with the CUS + Vehicle group for analysis of the long-term effects of ketamine (*p < .05). Nor were significant differences obtained in learning and memory performance in rats receiving ketamine (p > .05). CONCLUSION Taken together these findings demonstrate that a short-term administration of ketamine induced rapid antidepressant-like effects in adult male rats exposed to CUS conditions, effects that were not observed in response to the long-term treatment regime.
Collapse
Affiliation(s)
- Yinghong Jiang
- Department of PsychologyWeifang Medical UniversityShandongChina
| | - Yiqiang Wang
- School of Bioscience and TechnologyWeifang Medical UniversityShandongChina
| | - Xiaoran Sun
- Department of PsychologyWeifang Medical UniversityShandongChina
| | - Bo Lian
- School of Bioscience and TechnologyWeifang Medical UniversityShandongChina
| | - Hongwei Sun
- Department of PsychologyWeifang Medical UniversityShandongChina
| | - Gang Wang
- Laboratory for Cognitive NeuroscienceWeifang Medical UniversityShandongChina
| | - Zhongde Du
- Department of NeurologyChinese People's Liberation Army eighty‐nine HospitalShandongChina
| | - Qi Li
- Department of Psychiatry and Centre for Reproduction Growth and DevelopmentUniversity of Hong KongHongKongChina
| | - Lin Sun
- Department of PsychologyWeifang Medical UniversityShandongChina
- Laboratory for Cognitive NeuroscienceWeifang Medical UniversityShandongChina
| |
Collapse
|
50
|
Bartoli F, Riboldi I, Crocamo C, Di Brita C, Clerici M, Carrà G. Ketamine as a rapid-acting agent for suicidal ideation: A meta-analysis. Neurosci Biobehav Rev 2017; 77:232-236. [DOI: 10.1016/j.neubiorev.2017.03.010] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/09/2017] [Accepted: 03/17/2017] [Indexed: 11/16/2022]
|