1
|
Shi Y, Wu W. Advances in transcranial focused ultrasound neuromodulation for mental disorders. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111244. [PMID: 39756638 DOI: 10.1016/j.pnpbp.2024.111244] [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: 10/16/2024] [Revised: 12/29/2024] [Accepted: 12/29/2024] [Indexed: 01/07/2025]
Abstract
Mental disorders are a major public health concern, affecting millions worldwide. Current treatments have limitations, highlighting the need for novel, effective, and safe interventions. Transcranial focused ultrasound (tFUS), a non-invasive neuromodulation technology, has emerged as a promising tool for treating mental disorders due to its high controllability, precision, and safety. This review summarizes the research progress of tFUS in several major mental disorders, including depression, anxiety, schizophrenia, and substance use disorders (SUDs). Animal studies have demonstrated the efficacy of tFUS in improving psychiatric symptoms and modulating neural circuits through various mechanisms, such as enhancing neuronal activity, synaptic plasticity, and neurotransmitter release. Preliminary clinical trials have also shown the potential of tFUS in alleviating symptoms in patients with treatment-resistant mental disorders. Safety evaluation studies across in vitro, animal, and human levels have supported the overall safety of tFUS under commonly used parameters. tFUS has shown broad application prospects in treating mental disorders, supported by its efficacy in animal models and preliminary clinical trials. By modulating neuronal activity, synaptic plasticity, neurotransmitters, and brain networks, tFUS could improve psychiatric symptoms and regulate neural circuits. However, current research on tFUS in mental disorders is still in its early stages, and further studies are needed to elucidate its mechanisms of action, expand its applications, and conduct large-sample, long-term clinical trials to systematically evaluate its efficacy, protocol optimization, and safety. As an innovative neuromodulation technology, tFUS has the potential to complement conventional therapies and provide new hope for addressing the global challenge of mental disorders.
Collapse
Affiliation(s)
- Yu Shi
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Wen Wu
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| |
Collapse
|
2
|
Loiodice S, D'Acquisto F, Drinkenburg P, Suojanen C, Llorca PM, Manji HK. Neuropsychiatric drug development: Perspectives on the current landscape, opportunities and potential future directions. Drug Discov Today 2024; 30:104255. [PMID: 39615745 DOI: 10.1016/j.drudis.2024.104255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
Mental health represents a major challenge to our societies. One key difficulty associated with neuropsychiatric drug development is the lack of connection between the underlying biology and the disease. Nevertheless, there is growing optimism in this field with recent drug approvals (the first in decades) and renewed interest from pharmaceutical companies and investors. Here we review some of the most promising drug discovery and development endeavors currently deployed by industry. We also present elements illustrating the renewed interest from key stakeholders in neuropsychiatric drug development and provide potential future directions in this field.
Collapse
Affiliation(s)
| | - Fulvio D'Acquisto
- William Harvey Research Institute, School of Medicine and Dentistry, Queen Mary University of London, London, UK; School of Life and Health Science, University of Roehampton, London, UK
| | - Pim Drinkenburg
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Christian Suojanen
- Broadreach Global LLC, Miami, FL, USA; European Brain Council, Brussels, Belgium
| | - Pierre-Michel Llorca
- Department of Psychiatry, CHU Clermont-Ferrand, University of Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut Pascal (UMR 6602), Clermont-Ferrand, France; Fondation FondaMental, Créteil, France
| | - Husseini K Manji
- Oxford University, Oxford, UK; Yale University, New Haven, CT, USA; UK Government Mental Health Mission, London, UK
| |
Collapse
|
3
|
Gvozdeckii AN, Dobrovolskaya AE, Prokopovich GA, Sofronov AH. Efficacy and Safety Profiles of Antipsychotic Drugs as Viewed by Psychiatrists: A Comparative Analysis of Cariprazine and Risperidone. CONSORTIUM PSYCHIATRICUM 2023; 4:17-27. [PMID: 38618636 PMCID: PMC11009980 DOI: 10.17816/cp12049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 12/11/2023] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Physicians hold the belief that the treatment outcomes and the treatment strategy they eventually adopt is largely determined by the differences in medications. Despite numerous studies focusing on the decision-making processes of psychiatrists, including the choice of antipsychotics when prescribing pharmacotherapy, the impact of therapeutic drug profiling on physicians' decision-making remains poorly comprehended. AIM The aim of this study is to assess the quantitative differences in perceptions of antipsychotics by psychiatrists using cariprazine and risperidone as examples. METHODS A total of 79 psychiatrists were interviewed anonymously in St. Petersburg, Russia. The physicians documented the clinical advantages they perceived drugs to possess relative to one another, following a predetermined principle: A B, A=B, A B (2-AC protocol). The comparison is based on eleven parameters that assess the effectiveness and safety of cariprazine or risperidone. It has been hypothesized that the pattern of responses (qualitative difference) and the degree of preference for each drug (quantitative difference) may not align with the data in the original meta-analyses. RESULTS The perception parameter exhibited a greater difference than anticipated (δ - 0.889), while the threshold for differentiating between the drugs was lower (τ - 1.001). The response pattern only aligned with theory by 44.37%. The dispersion of responses was associated with the length of work experience. CONCLUSION The perceived difference between the drugs significantly deviates from the theoretical data, both in terms of strength of perception and pattern (quantitative and qualitative differences).
Collapse
|
4
|
Poian LR, Chiavegatto S. Serotonin Syndrome: The Role of Pharmacology in Understanding Its Occurrence. Cureus 2023; 15:e38897. [PMID: 37309350 PMCID: PMC10257984 DOI: 10.7759/cureus.38897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/14/2023] Open
Abstract
Serotonin syndrome (SS) is a potentially fatal adverse drug reaction characterized by an exaggerated increase in serotonergic activity in the central and peripheral nervous systems. It presents a constellation of signs and symptoms related to behavioral changes, neuromuscular excitability, and autonomic instability. These symptoms can occur in both mild and severe forms. SS can be triggered by the therapeutic use of a drug that increases serotonin (5-HT) availability in the synaptic cleft or by the co-administration of two or more drugs that provide this increase. With the escalating use of antidepressants by the world's population, this adverse reaction may be more recurrent. However, SS is often overlooked by patients or not diagnosed by doctors. This review aims to improve awareness about SS and provide a pharmacological perspective to explain its occurrence. Evidence shows that other neurotransmitters may also be involved with the pathology of SS. Furthermore, SS and neuroleptic malignant syndrome (NMS) seem to be part of the same pathological spectrum, especially in atypical NMS cases. The emergence of the syndrome's symptoms may be closely related to pharmacokinetic and/or pharmacodynamic polymorphisms that lead to an increase in the 5-HT available to or 5-HT signaling by specific receptors, thus constituting an important area for future investigations.
Collapse
Affiliation(s)
- Leila R Poian
- Department of Pharmacology, Biomedical Sciences Institute, University of Sao Paulo (ICB-USP), Sao Paulo, BRA
| | - Silvana Chiavegatto
- Department of Pharmacology, Biomedical Sciences Institute, University of Sao Paulo (ICB-USP), Sao Paulo, BRA
- Department of Psychiatry, Institute of Psychiatry, University of Sao Paulo Medical School (FMUSP), Sao Paulo, BRA
| |
Collapse
|
5
|
de Filippis R, Staltari FA, Aloi M, Carbone EA, Rania M, Destefano L, Steardo Jr. L, Segura-Garcia C, De Fazio P. Effectiveness of SGA-LAIs on Clinical, Cognitive, and Social Domains in Schizophrenia: Results from a Prospective Naturalistic Study. Brain Sci 2023; 13:brainsci13040577. [PMID: 37190542 DOI: 10.3390/brainsci13040577] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/21/2023] [Accepted: 03/26/2023] [Indexed: 03/31/2023] Open
Abstract
We hypothesized that shifting from oral second-generation antipsychotics (SGA) to their long-acting injectable (LAI) counterpart would be beneficial for the psychopathological, cognitive, social, and general health domains in outpatients suffering from schizophrenia. We aimed to evaluate the prospective usefulness of SGA-LAI treatment by carrying out a head-to-head comparison of two different medications (i.e., aripiprazole monohydrate (Ari-LAI) and paliperidone palmitate 1 and 3 month (PP1M, PP3M)) in a real-world setting, assessing the effectiveness and tolerability of Ari-LAI and PP1M/PP3M over a 15 month follow-up. A total of 69 consecutive individuals affected by schizophrenia were screened for eligibility. Finally, 46 outpatients (29 treated with Ari-LAI, 13 with PP1M, and four with PP3M) were evaluated through clinical, functional, and neuropsychological assessment administrated at baseline and after 3-, 12-, and 15-month follow-up periods. Moreover, periodic general medical evaluations were carried out. We estimated an overall improvement over time on the explored outcomes, without differences with respect to the type of LAI investigated, and with a global 16.4% dropout rate. Our findings suggest that switching from oral SGA to SGA-LAIs represents a valid and effective treatment strategy, with significant improvements on psychopathological, cognitive, social, and clinical variables for patients suffering from schizophrenia, regardless of the type of molecule chosen.
Collapse
|
6
|
Rahman MM, Islam MR, Alam Tumpa MA, Shohag S, Shakil Khan Shuvo, Ferdous J, Kajol SA, Aljohani ASM, Al Abdulmonem W, Rauf A, Thiruvengadam M. Insights into the promising prospect of medicinal chemistry studies against neurodegenerative disorders. Chem Biol Interact 2023; 373:110375. [PMID: 36739931 DOI: 10.1016/j.cbi.2023.110375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/06/2022] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Medicinal chemistry is an interdisciplinary field that incorporates organic chemistry, biochemistry, physical chemistry, pharmacology, informatics, molecular biology, structural biology, cell biology, and other disciplines. Additionally, it considers molecular factors such as the mode of action of the drugs, their chemical structure-activity relationship (SAR), and pharmacokinetic aspects like absorption, distribution, metabolism, elimination, and toxicity. Neurodegenerative disorders (NDs), which are defined by the breakdown of neurons over time, are affecting an increasing number of people. Oxidative stress, particularly the increased production of Reactive Oxygen Species (ROS), plays a crucial role in the growth of various disorders, as indicated by the identification of protein, lipid, and Deoxyribonucleic acid (DNA) oxidation products in vivo. Because of their inherent nature, most biological molecules are vulnerable to ROS, even if they play a role in metabolic parameters and cell signaling. Due to their high polyunsaturated fatty acid content, low antioxidant barrier, and high oxygen uptake, neurons are particularly vulnerable to oxidation by nature. As a result, excessive ROS generation in neurons looks especially harmful, and the mechanisms associated with biomolecule oxidative destruction are several and complex. This review focuses on the formation and management of ROS, as well as their chemical characteristics (both thermodynamic and kinetic), interactions, and implications in NDs.
Collapse
Affiliation(s)
- Md Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Mst Afroza Alam Tumpa
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Sheikh Shohag
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University Buraydah, 52571, Saudi Arabia
| | - Shakil Khan Shuvo
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Jannatul Ferdous
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Saima Akter Kajol
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University Buraydah, 52571, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine Qassim University, Buraydah, Saudi Arabia
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, 23430, Khyber Pakhtunkhwa (KP), Pakistan.
| | - Muthu Thiruvengadam
- Department of Applied Bioscience, College of Life and Environmental Sciences, Konkuk University, Seoul, 05029, South Korea; Department of Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India.
| |
Collapse
|
7
|
Crismon ML, Walkow J, Sommi RW. Drug Development for New Psychiatric Drug Therapies. ADVANCES IN NEUROBIOLOGY 2023; 30:131-167. [PMID: 36928848 DOI: 10.1007/978-3-031-21054-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Drug development is an expensive, high risk, and highly regulated process. Only about 6.2% of new molecules tested for mental disorders eventually achieve Food and Drug Administration (FDA) approval. New molecular entities are produced, and extensive in vitro animal testing is performed before they are evaluated in humans. The compound is used in animals to predict clinical effects in humans, and studies addressing pharmacodynamics, pharmacokinetics, toxicology, and mutagenicity are conducted. Human research proceeds in three stages with the ultimate goal of proving that a new agent is efficacious and safe for a treatment of a specific disease in humans. If efficacy and safety are demonstrated in two Phase III studies, then the sponsor can submit a new drug application (NDA) to the FDA. The FDA oversees each step of the process to assure that good research practices are followed, data integrity is assured, and human research subjects are protected.
Collapse
Affiliation(s)
| | - Janet Walkow
- The University of Texas at Austin, Austin, TX, USA
| | - Roger W Sommi
- University of Missouri at Kansas City, Kansas City, MO, USA
| |
Collapse
|
8
|
Wang Y, Liu Z, Lu J, Wang W, Wang L, Yang Y, Wang H, Ye L, Zhang J, Tian J. Biological evaluation and in silico studies of novel compounds as potent TAAR1 agonists that could be used in schizophrenia treatment. Front Pharmacol 2023; 14:1161964. [PMID: 37153799 PMCID: PMC10160475 DOI: 10.3389/fphar.2023.1161964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/05/2023] [Indexed: 05/10/2023] Open
Abstract
Introduction: Schizophrenia is a serious mental illness that requires effective treatment with minimal adverse effects. As preclinical and clinical research progresses, trace amine-associated receptor 1 (TAAR1) is becoming a potential new target for the treatment of schizophrenia. Methods: We used molecular docking and molecular dynamics (MD) simulations to discover TAAR1 agonists. The agonistic or inhibitory effects of compounds on TAAR1, 5-HT1A, 5-HT2A, and dopamine D2-like receptors were determined. We used an MK801-induced schizophrenia-like behavior model to assess the potential antipsychotic effects of compounds. We also performed a catalepsy assay to detect the adverse effects. To evaluate the druggability of the compounds, we conducted evaluations of permeability and transporter substrates, liver microsomal stability in vitro, human ether-à-go-go-related gene (hERG), pharmacokinetics, and tissue distribution. Results: We discovered two TAAR1 agonists: compounds 50A and 50B. The latter had high TAAR1 agonistic activity but no agonistic effect on dopamine D2-like receptors and demonstrated superior inhibition of MK801-induced schizophrenia-like behavior in mice. Interestingly, 50B had favorable druggability and the ability to penetrate the blood-brain barrier (BBB) without causing extrapyramidal symptoms (EPS), such as catalepsy in mice. Conclusion: These results demonstrate the potential beneficial role of TAAR1 agonists in the treatment of schizophrenia. The discovery of a structurally novel TAAR1 agonist (50B) may provide valuable assistance in the development of new treatments for schizophrenia.
Collapse
Affiliation(s)
- Yunjie Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Zhaofeng Liu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Jing Lu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Wenyan Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Lin Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Yifei Yang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Liang Ye
- School of Public Health and Management, Binzhou Medical University, Yantai, China
- *Correspondence: Liang Ye, ; Jianzhao Zhang, ; Jingwei Tian,
| | - Jianzhao Zhang
- College of Life Sciences, Yantai University, Yantai, China
- *Correspondence: Liang Ye, ; Jianzhao Zhang, ; Jingwei Tian,
| | - Jingwei Tian
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
- *Correspondence: Liang Ye, ; Jianzhao Zhang, ; Jingwei Tian,
| |
Collapse
|
9
|
TAAR1 dependent and independent actions of the potential antipsychotic and dual TAAR1/5-HT 1A receptor agonist SEP-383856. Neuropsychopharmacology 2022; 47:2319-2329. [PMID: 36100653 PMCID: PMC9630386 DOI: 10.1038/s41386-022-01421-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 07/04/2022] [Accepted: 07/30/2022] [Indexed: 11/09/2022]
Abstract
SEP-383856 (SEP-856) is a novel antipsychotic under clinical development. It displays a unique pattern of receptor interaction, with only weak (partial agonist) activity at dopamine D2 receptors, yet more potent agonist activity at the trace amine associated receptor (TAAR1) and 5-hydroxytryptamine 1 A receptor (5-HT1A). Nonetheless, these observations await independent confirmation and more detailed characterization of the in vitro and in vivo actions of SEP-856 at TAAR1 and 5-HT1A receptors would be instructive. Herein, we employed luminescence complementation technology in heterologous live cell systems, confocal microscopy, voltage clamp electrophysiology, behavioral readouts and TAAR1 knockout (KO) mice to study SEP-856 in further detail. We provide evidence for the ability of SEP-856 to activate TAAR1 at the surface plasma membrane, and show that this interaction results in Gαs recruitment (pEC50: 6.08 ± 0.22 EMAX: 96.41% ± 15.26) and by extension, to G-protein inwardly rectifying potassium (GIRK) channel activation. Using TAAR1-KO mice, we find TAAR1 to be indispensable for SEP-856 control of body temperature, baseline locomotion reduction and for "antipsychotic-like" efficacy as characterized by a reversal of dizocilipine (MK-801) mediated disruption of pre-pulse inhibition. Conversely, the inhibition by SEP-856 of MK-801 induced locomotion was unaffected in TAAR1 KO mice. SEP-856 behaved as a low-potency, partial agonist at the 5-HT1A receptor, while it partially inhibited recruitment of D2 receptor-coupled Gα and GIRK by DA and acted as a weak partial agonist with low potency at the same receptor when applied alone. Our findings corroborate and extend previous observations on the molecular substrates engaged by this unique, dual TAAR1/5-HT1A receptor agonist and potential antipsychotic that could prove to have major advantages in the treatment of schizophrenia and other psychotic disorders.
Collapse
|
10
|
Mathematical Model of Interaction of Therapist and Patients with Bipolar Disorder: A Systematic Literature Review. J Pers Med 2022; 12:jpm12091469. [PMID: 36143254 PMCID: PMC9503456 DOI: 10.3390/jpm12091469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/04/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Mood swings in patients with bipolar disorder (BD) are difficult to control and can lead to self-harm and suicide. The interaction between the therapist and BD will determine the success of therapy. The interaction model between the therapist and BD begins by reviewing the models that were previously developed using the Systematic Literature Review and Bibliometric methods. The limit of articles used was sourced from the Science Direct, Google Scholar, and Dimensions databases from 2009 to 2022. The results obtained were 67 articles out of a total of 382 articles, which were then re-selected. The results of the selection of the last articles reviewed were 52 articles. Using VOSviewer version 1.6.16, a visualization of the relationship between the quotes “model”, “therapy”, “emotions”, and “bipolar disorder” can be seen. This study also discusses the types of therapy that can be used by BD, as well as treatment innovations and the mathematical model of the therapy itself. The results of this study are expected to help further researchers to develop an interaction model between therapists and BD to improve the quality of life of BD.
Collapse
|
11
|
Harrison PJ, Mould A, Tunbridge EM. New drug targets in psychiatry: Neurobiological considerations in the genomics era. Neurosci Biobehav Rev 2022; 139:104763. [PMID: 35787892 DOI: 10.1016/j.neubiorev.2022.104763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/15/2022] [Accepted: 06/14/2022] [Indexed: 01/11/2023]
Abstract
After a period of withdrawal, pharmaceutical companies have begun to reinvest in neuropsychiatric disorders, due to improvements in our understanding of these disorders, stimulated in part by genomic studies. However, translating this information into disease insights and ultimately into tractable therapeutic targets is a major challenge. Here we consider how different sources of information might be integrated to guide this process. We review how an understanding of neurobiology has been used to advance therapeutic candidates identified in the pre-genomic era, using catechol-O-methyltransferase (COMT) as an exemplar. We then contrast with ZNF804A, the first genome-wide significant schizophrenia gene, and draw on some of the lessons that these and other examples provide. We highlight that, at least in the short term, the translation of potential targets for which there is orthogonal neurobiological support is likely to be more straightforward and productive than that those relying solely on genomic information. Although we focus here on information from genomic studies of schizophrenia, the points are broadly applicable across major psychiatric disorders and their symptoms.
Collapse
Affiliation(s)
- Paul J Harrison
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Arne Mould
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Elizabeth M Tunbridge
- Department of Psychiatry, University of Oxford, Oxford, UK; Oxford Health NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
12
|
Krishnamurthy N, Grimshaw AA, Axson SA, Choe SH, Miller JE. Drug repurposing: a systematic review on root causes, barriers and facilitators. BMC Health Serv Res 2022; 22:970. [PMID: 35906687 PMCID: PMC9336118 DOI: 10.1186/s12913-022-08272-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Repurposing is a drug development strategy receiving heightened attention after the Food and Drug Administration granted emergency use authorization of several repurposed drugs to treat Covid-19. There remain knowledge gaps on the root causes, facilitators and barriers for repurposing. METHOD This systematic review used controlled vocabulary and free text terms to search ABI/Informa, Academic Search Premier, Business Source Complete, Cochrane Library, EconLit, Google Scholar, Ovid Embase, Ovid Medline, Pubmed, Scopus, and Web of Science Core Collection databases for the characteristics, reasons and example of companies deprioritizing development of promising drugs and barriers, facilitators and examples of successful re-purposing. RESULTS We identified 11,814 articles, screened 5,976 for relevance, found 437 eligible for full text review, 115 of which were included in full analysis. Most articles (66%, 76/115) discussed why promising drugs are abandoned, with lack of efficacy or superiority to other therapies (n = 59), strategic business reasons (n = 35), safety problems (n = 28), research design decisions (n = 12), the complex nature of a studied disease or drug (n = 7) and regulatory bodies requiring more information (n = 2) among top reasons. Key barriers to repurposing include inadequate resources (n = 42), trial data access and transparency around abandoned compounds (n = 20) and expertise (n = 11). Additional barriers include uncertainty about the value of repurposing (n = 13), liability risks (n = 5) and intellectual property (IP) challenges (n = 26). Facilitators include the ability to form multi-partner collaborations (n = 38), access to compound databases and database screening tools (n = 32), regulatory modifications (n = 5) and tax incentives (n = 2). CONCLUSION Promising drugs are commonly shelved due to insufficient efficacy or superiority to alternate therapies, poor market prospects, and industry consolidation. Inadequate resources and data access and challenges negotiating IP are key barriers to repurposing reaching its full potential as a core approach in drug development. Multi-partner collaborations and the availability and use of compound databases and tax incentives are key facilitators for repurposing. More research is needed on the current value of repurposing in drug development and how to better facilitate resources to support it, where valuable, especially financial, staffing for out-licensing shelved products, and legal expertise to negotiate IP agreements in multi-partner collaborations. TRIAL REGISTRATION The protocol was registered on Open Science Framework ( https://osf.io/f634k/ ) as it was not eligible for registration on PROSPERO as the review did not focus on a health-related outcome.
Collapse
Affiliation(s)
- Nithya Krishnamurthy
- Internal Medicine Department, Yale University School of Medicine, 367 Cedar Street, 4th Floor, New Haven, CT, 06520, USA
| | - Alyssa A Grimshaw
- Cushing/Whitney Medical Library, Yale University, 333 Cedar Street, Box 208014, New Haven, CT, 06520, USA
| | - Sydney A Axson
- Internal Medicine Department, Yale University School of Medicine, 367 Cedar Street, 4th Floor, New Haven, CT, 06520, USA
| | - Sung Hee Choe
- Milken Institute Center for Faster Cures, 730 15th Street NW, Washington, DC, 20005, USA
| | - Jennifer E Miller
- Internal Medicine Department, Yale University School of Medicine, 367 Cedar Street, 4th Floor, New Haven, CT, 06520, USA.
| |
Collapse
|
13
|
Zhu S, Liu F, Zhang R, Xiong Z, Zhang Q, Hao L, Chen S. Neuroprotective Potency of Neolignans in Magnolia officinalis Cortex Against Brain Disorders. Front Pharmacol 2022; 13:857449. [PMID: 35784755 PMCID: PMC9244706 DOI: 10.3389/fphar.2022.857449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/20/2022] [Indexed: 12/03/2022] Open
Abstract
In recent years, neurological diseases including Alzheimer’s disease, Parkinson’s disease and stroke are one of the main causes of death in the world. At the same time, the incidence of psychiatric disorders including depression and anxiety has been increasing. Accumulating elderly and stressed people suffer from these brain disorders, which is undoubtedly a huge burden on the modern aging society. Neolignans, the main active ingredients in Magnolia officinalis cortex, were reported to have neuroprotective effects. In addition, the key bioactive ingredients of neolignans, magnolol (1) and honokiol (2), were proved to prevent and treat neurological diseases and psychiatric disorders by protecting nerve cells and brain microvascular endothelial cells (BMECs). Furthermore, neolignans played a role in protecting nerve cells via regulation of neuronal function, suppression of neurotoxicity, etc. This review summarizes the neuroprotective effect, primary mechanisms of the leading neolignans and provides new prospects for the treatment of brain disorders in the future.
Collapse
Affiliation(s)
- Shun Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fang Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Fang Liu, ; Shiyin Chen,
| | - Ruiyuan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zongxiang Xiong
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Hao
- Huarun Sanjiu (ya’an) Pharmaceutical Group Co., LTD., Ya’an, China
| | - Shiyin Chen
- Department of Orthopedics of Traditional Chinese Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Fang Liu, ; Shiyin Chen,
| |
Collapse
|
14
|
Wium-Andersen MK, Jørgensen TSH, Jørgensen MB, Rungby J, Hjorthøj C, Sørensen HJ, Osler M. The association between birth weight, ponderal index, psychotropic medication, and type 2 diabetes in individuals with severe mental illness. J Diabetes Complications 2022; 36:108181. [PMID: 35346563 DOI: 10.1016/j.jdiacomp.2022.108181] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/27/2022] [Accepted: 03/14/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Impaired fetal growth may increase vulnerability towards metabolic disturbances associated with some medications. We examined whether birth weight and ponderal index modify the association between psychotropic medication and type 2 diabetes among young adults with severe psychiatric diagnosis. METHODS A total of 36,957 individuals born in Denmark between 1973 and 1983 with a diagnosis of schizophrenia, bipolar disorder, or depression were followed from first diagnosis until 2018. Cox proportional hazard models were applied to analyse risk of type 2 diabetes with use of psychotropic medications and interactions between psychotropic medication and birth weight and ponderal index, respectively. RESULTS During follow-up, 1575 (4.2%) individuals received a diagnosis of type 2 diabetes. Use of antipsychotic, mood stabilizing and antidepressant medications were associated with higher hazard ratios (HRs) of type 2 diabetes (HRantipsychotics 1.68 [95%CI 1.49-1.90]; HRmood stabilizing medication 1.41 [95%CI 1.25-1.59]; HRantidepressants 2.00 [95%CI 1.68-2.37]), as were a birth weight below 2500 g (HR 1.13 [95%CI 1.01-1.28]), and high ponderal index (HR 1.26 [95%CI 1.11-1.43]). The highest rates of type 2 diabetes for each psychotropic medication category were found in medication users with low birth weight or high ponderal index. However, neither birth weight nor ponderal index significantly modified the association between psychotropic medication and diabetes risk. CONCLUSION Psychotropic medication use, birth weight, and ponderal index were risk factors for type 2 diabetes in patients with severe mental illness, but neither birth weight nor ponderal index modified the association between psychotropic medication and type 2 diabetes.
Collapse
Affiliation(s)
| | - Terese Sara Høj Jørgensen
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Denmark; Department of Public Health, Section of Social Medicine, University of Copenhagen, Denmark
| | - Martin Balslev Jørgensen
- Psychiatric Center Copenhagen, Rigshospitalet, Denmark; Institute of Clinical Medicine, University of Copenhagen, Denmark
| | - Jørgen Rungby
- Department of Endocrinology and Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Denmark
| | - Carsten Hjorthøj
- Copenhagen Research Center for Mental Health-CORE, Mental Health Center Copenhagen, Copenhagen University Hospital, Denmark; Department of Public Health, Section of Epidemiology, University of Copenhagen, Denmark
| | - Holger J Sørensen
- Copenhagen Research Center for Mental Health-CORE, Mental Health Center Copenhagen, Copenhagen University Hospital, Denmark
| | - Merete Osler
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Denmark; Department of Public Health, Section of Epidemiology, University of Copenhagen, Denmark.
| |
Collapse
|
15
|
Gurtner C, Lohrmann C, Schols JMGA, Hahn S. Shared Decision Making in the Psychiatric Inpatient Setting: An Ethnographic Study about Interprofessional Psychiatric Consultations. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19063644. [PMID: 35329331 PMCID: PMC8954628 DOI: 10.3390/ijerph19063644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/11/2022] [Accepted: 03/16/2022] [Indexed: 11/29/2022]
Abstract
Shared decision making is increasingly receiving attention in health care and might improve both the quality of care and patient outcomes. Nevertheless, due to its complexity, implementation of shared decision making in clinical practice seems challenging. This ethnographic study aimed to gain a better understanding of how psychiatric inpatients and the interprofessional care team interact during regular interprofessional psychiatric consultations. Data were collected through participant observation on two different psychiatric wards in a large psychiatric hospital in Switzerland. The observation focused on the contextual aspects of interprofessional patient consultations, the communication and interaction as well as the extent to which patients were involved in decision making. Participants included patients, psychiatrists, junior physicians, nurses, psychologists, social workers and therapists. We observed 71 interprofessional psychiatric consultations and they differed substantially in both wards in terms of context (place and form) and culture (way of interacting). On the contrary, results showed that the level of patient involvement in decision making was comparable and depended on individual factors, such as the health care professionals’ communication style as well as the patients’ personal initiative to be engaged. The main topics discussed with the patients related to pharmacotherapy and patient reported symptoms. Health care professionals in both wards used a rather unidirectional communication style. Therefore, in order to promote patient involvement in the psychiatric inpatient setting, rather than to focus on contextual factors, consultations should follow a specific agenda and promoting a bidirectional communication style for all parties involved is strongly recommended.
Collapse
Affiliation(s)
- Caroline Gurtner
- Applied Research and Development in Nursing, Department of Health Professions, Bern University of Applied Sciences, 3008 Bern, Switzerland;
- Department of Health Services Research, Care and Public Health Research Institute (CAPHRI), Maastricht University, 6200 MD Maastricht, The Netherlands;
- Correspondence:
| | - Christa Lohrmann
- Institute of Nursing Science, Medical University Graz, 8010 Graz, Austria;
| | - Jos M. G. A. Schols
- Department of Health Services Research, Care and Public Health Research Institute (CAPHRI), Maastricht University, 6200 MD Maastricht, The Netherlands;
- Department of Family Medicine & Care and Public Health Research Institute (CAPHRI), Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Sabine Hahn
- Applied Research and Development in Nursing, Department of Health Professions, Bern University of Applied Sciences, 3008 Bern, Switzerland;
| |
Collapse
|
16
|
Costi S, Han MH, Murrough JW. The Potential of KCNQ Potassium Channel Openers as Novel Antidepressants. CNS Drugs 2022; 36:207-216. [PMID: 35258812 DOI: 10.1007/s40263-021-00885-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2021] [Indexed: 12/12/2022]
Abstract
Major depressive disorder (MDD) is a leading cause of disability worldwide and less than one-third of patients with MDD achieve stable remission of symptoms, despite currently available treatments. Although MDD represents a serious health problem, a complete understanding of the neurobiological mechanisms underlying this condition continues to be elusive. Accumulating evidence from preclinical and animal studies provides support for the antidepressant potential of modulators of KCNQ voltage-gated potassium (K+) channels. KCNQ K+ channels, through regulation of neuronal excitability and activity, contribute to neurophysiological mechanisms underlying stress resilience, and represent potential targets of drug discovery for depression. The present article focuses on the pharmacology and efficacy of KCNQ2/3 K+ channel openers as novel therapeutic agents for depressive disorders from initial studies conducted on animal models showing depressive-like behaviors to recent work in humans that examines the potential for KCNQ2/3 channel modulators as novel antidepressants. Data from preclinical work suggest that KCNQ-type K+ channels are an active mediator of stress resilience and KCNQ2/3 K+ channel openers show antidepressant efficacy. Similarly, evidence from clinical trials conducted in patients with MDD using the KCNQ2/3 channel opener ezogabine (retigabine) showed significant improvements in depressive symptoms and anhedonia. Overall, KCNQ channel openers appear a promising target for the development of novel therapeutics for the treatment of psychiatric disorders and specifically for MDD.
Collapse
Affiliation(s)
- Sara Costi
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA
| | - Ming-Hu Han
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Center for Affective Neuroscience, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - James W Murrough
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA. .,Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
17
|
Cigognini MA, Guirado AG, van de Meene D, Schneider MA, Salomon MS, de Alexandria VS, Adriano JP, Thaler AM, Fernandes FDS, Carneiro A, Moreno RA. Intramuscular ketamine vs. escitalopram and aripiprazole in acute and maintenance treatment of patients with treatment-resistant depression: A randomized double-blind clinical trial. Front Psychiatry 2022; 13:830301. [PMID: 35935445 PMCID: PMC9354749 DOI: 10.3389/fpsyt.2022.830301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Ketamine, an N-methyl D-aspartate (NMDA) receptor antagonist, can promote rapid action in the management of individuals with treatment-resistant depression (TRD) at sub-anesthetic doses. However, few studies have investigated the long-term use of ketamine administered intravenously (IV) and intranasally (IN). We report the design and rationale of a therapeutic trial for assessing the efficacy, safety, and tolerability of repeated-dose intramuscular (IM) ketamine vs. active treatment (escitalopram and aripiprazole) in TRD patients. METHODS A comparative, parallel-group, randomized double-blind trial assessing the efficacy, safety, and tolerability of acute (4 weeks) and maintenance (24 weeks) use of IM ketamine (0.75 mg/kg) vs. active control (escitalopram 15 mg and aripiprazole 5 mg) in individuals with moderate-severe intensity TRD (no psychotic symptoms) with or without suicide risk will be conducted. Patients with TRD (18-40 years) will be randomized and blinded to receive ketamine IM or active treatment at a 1:1 ratio for 4 weeks (active treatment) and 24 weeks (maintenance treatment). Subjects will be assessed using clinical scales, monitored for vital signs (VS) after application of injectable medication, and undergo neuropsychological tests. The primary outcome will be changed on the Montgomery-Åsberg Depression Rating Scale (MADRS) during the course of the trial. The study is in running. RESULTS This study can potentially yield evidence on the use of IM ketamine in the treatment of depressive disorders as an ultra-rapid low-cost therapy associated with less patient discomfort and reduced use of medical resources, and can elucidate long-term effects on different outcomes, such as neuropsychological aspects. CONCLUSIONS The trial can help promote the introduction of a novel accessible approach for the treatment of complex disease (TRD) and also allow refinement of its long-term use. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ct2/show/NCT04234776, identifier: NCT04234776.
Collapse
Affiliation(s)
- Marco Aurélio Cigognini
- Mood Disorders Unit (GRUDA), Institute and Department of Psychiatry, School of Medicine, University of São Paulo (FMUSP), São Paulo, Brazil
| | - Alia Garrudo Guirado
- Mood Disorders Unit (GRUDA), Institute and Department of Psychiatry, School of Medicine, University of São Paulo (FMUSP), São Paulo, Brazil
| | - Denise van de Meene
- Mood Disorders Unit (GRUDA), Institute and Department of Psychiatry, School of Medicine, University of São Paulo (FMUSP), São Paulo, Brazil
| | - Mônica Andréia Schneider
- Mood Disorders Unit (GRUDA), Institute and Department of Psychiatry, School of Medicine, University of São Paulo (FMUSP), São Paulo, Brazil
| | - Mônica Sarah Salomon
- Mood Disorders Unit (GRUDA), Institute and Department of Psychiatry, School of Medicine, University of São Paulo (FMUSP), São Paulo, Brazil
| | - Vinicius Santana de Alexandria
- Mood Disorders Unit (GRUDA), Institute and Department of Psychiatry, School of Medicine, University of São Paulo (FMUSP), São Paulo, Brazil
| | - Juliana Pisseta Adriano
- Mood Disorders Unit (GRUDA), Institute and Department of Psychiatry, School of Medicine, University of São Paulo (FMUSP), São Paulo, Brazil
| | - Ana Maria Thaler
- Mood Disorders Unit (GRUDA), Institute and Department of Psychiatry, School of Medicine, University of São Paulo (FMUSP), São Paulo, Brazil
| | - Fernando Dos Santos Fernandes
- Mood Disorders Unit (GRUDA), Institute and Department of Psychiatry, School of Medicine, University of São Paulo (FMUSP), São Paulo, Brazil
| | - Adriana Carneiro
- Mood Disorders Unit (GRUDA), Institute and Department of Psychiatry, School of Medicine, University of São Paulo (FMUSP), São Paulo, Brazil
| | - Ricardo Alberto Moreno
- Mood Disorders Unit (GRUDA), Institute and Department of Psychiatry, School of Medicine, University of São Paulo (FMUSP), São Paulo, Brazil
| |
Collapse
|
18
|
Riesel A, Endrass T, Weinberg A. Biomarkers of mental disorders: Psychophysiological measures as indicators of mechanisms, risk, and outcome prediction. Int J Psychophysiol 2021; 168:21-26. [PMID: 34364039 DOI: 10.1016/j.ijpsycho.2021.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Anja Riesel
- University of Hamburg, Department of Clinical Psychology and Psychotherapy, Germany.
| | - Tanja Endrass
- Technische Universität Dresden, Faculty of Psychology, Institute of Clinical Psychology and Psychotherapy, Addiction Research, Germany
| | | |
Collapse
|
19
|
Cadoná FC, de Souza DV, Fontana T, Bodenstein DF, Ramos AP, Sagrillo MR, Salvador M, Mota K, Davidson CB, Ribeiro EE, Andreazza AC, Machado AK. Açaí (Euterpe oleracea Mart.) as a Potential Anti-neuroinflammatory Agent: NLRP3 Priming and Activating Signal Pathway Modulation. Mol Neurobiol 2021; 58:4460-4476. [PMID: 34021869 DOI: 10.1007/s12035-021-02394-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/13/2021] [Indexed: 12/20/2022]
Abstract
Neurological disorders have been demonstrated to be associated with mitochondrial dysfunction. This impairment may lead to oxidative stress and neuroinflammation, specifically promoted by NLRP3 expression. Açaí (Euterpe oleracea Mart.) has been studied in this field, since it presents important biological activities. We investigated açaí extract's anti-neuroinflammatory capacity, through NLRP3 inflammasome modulation. Microglia (EOC 13.31) were exposed to LPS and nigericin, as agents of inflammatory induction, and treated with açaí extract. Additionally, we used lithium (Li) as an anti-inflammatory control. Three different experiment models were conducted: (1) isolated NLRP3 priming and activation signals; (2) combined NLRP3 priming and activation signals followed by açaí extract as a therapeutic agent; and (3) combined NLRP3 priming and activation signals with açaí extract as a preventive agent. Cells exposed to 0.1 µg/mL of LPS presented high proliferation and increased levels of NO, and ROS, while 0.1 µg/mL of açaí extract was capable to reduce cellular proliferation and recover levels of NO and ROS. Primed and activated cells presented increased levels of NLRP3, caspase-1, and IL-1β, while açaí, Li, and orientin treatments reversed this impairment. We found that açaí, Li, and orientin were effective prophylactic treatments. Preventative treatment with Li and orientin was unable to avoid overexpression of IL-1β compared to the positive control. However, orientin downregulated NLRP3 and caspase-1. Lastly, primed and activated cells impaired ATP production, which was prevented by pre-treatment with açaí, Li, and orientin. In conclusion, we suggest that açaí could be a potential agent to treat or prevent neuropsychiatric diseases related to neuroinflammation.
Collapse
Affiliation(s)
- Francine Carla Cadoná
- Graduate Program in Health and Life Sciences, Franciscan University, Santa Maria, RS, Brazil
| | - Diulie Valente de Souza
- Graduate Program in Nanoscience, Franciscan University, Santa Maria, RS, Brazil
- Laboratory of Cell Culture and Genetics, Franciscan University, Santa Maria, RS, Brazil
| | - Tuyla Fontana
- Laboratory of Cell Culture and Genetics, Franciscan University, Santa Maria, RS, Brazil
| | - David Frederick Bodenstein
- Department of Pharmacology and Toxicology, University of Toronto, Medical Science Building, Room 4211, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | | | | | - Mirian Salvador
- Biotechnology Institute, University of Caxias do Sul, Caxias do Sul, RS, Brazil
| | - Kennya Mota
- Third Age Open University Foundation, University of Amazonas State, Manaus, AM, Brazil
| | | | - Euler Esteves Ribeiro
- Third Age Open University Foundation, University of Amazonas State, Manaus, AM, Brazil
| | - Ana Cristina Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Medical Science Building, Room 4211, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
| | - Alencar Kolinski Machado
- Graduate Program in Nanoscience, Franciscan University, Santa Maria, RS, Brazil
- Laboratory of Cell Culture and Genetics, Franciscan University, Santa Maria, RS, Brazil
| |
Collapse
|
20
|
Costi S, Morris LS, Kirkwood KA, Hoch M, Corniquel M, Vo-Le B, Iqbal T, Chadha N, Pizzagalli DA, Whitton A, Bevilacqua L, Jha MK, Ursu S, Swann AC, Collins KA, Salas R, Bagiella E, Parides MK, Stern ER, Iosifescu DV, Han MH, Mathew SJ, Murrough JW. Impact of the KCNQ2/3 Channel Opener Ezogabine on Reward Circuit Activity and Clinical Symptoms in Depression: Results From a Randomized Controlled Trial. Am J Psychiatry 2021; 178:437-446. [PMID: 33653118 PMCID: PMC8791195 DOI: 10.1176/appi.ajp.2020.20050653] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Preclinical studies point to the KCNQ2/3 potassium channel as a novel target for the treatment of depression and anhedonia, a reduced ability to experience pleasure. The authors conducted the first randomized placebo-controlled trial testing the effect of the KCNQ2/3 positive modulator ezogabine on reward circuit activity and clinical outcomes in patients with depression. METHODS Depressed individuals (N=45) with elevated levels of anhedonia were assigned to a 5-week treatment period with ezogabine (900 mg/day; N=21) or placebo (N=24). Participants underwent functional MRI during a reward flanker task at baseline and following treatment. Clinical measures of depression and anhedonia were collected at weekly visits. The primary endpoint was the change from baseline to week 5 in ventral striatum activation during reward anticipation. Secondary endpoints included depression and anhedonia severity as measured using the Montgomery-Åsberg Depression Rating Scale (MADRS) and the Snaith-Hamilton Pleasure Scale (SHAPS), respectively. RESULTS The study did not meet its primary neuroimaging endpoint. Participants in the ezogabine group showed a numerical increase in ventral striatum response to reward anticipation following treatment compared with participants in the placebo group from baseline to week 5. Compared with placebo, ezogabine was associated with a significantly larger improvement in MADRS and SHAPS scores and other clinical endpoints. Ezogabine was well tolerated, and no serious adverse events occurred. CONCLUSIONS The study did not meet its primary neuroimaging endpoint, although the effect of treatment was significant on several secondary clinical endpoints. In aggregate, the findings may suggest that future studies of the KCNQ2/3 channel as a novel treatment target for depression and anhedonia are warranted.
Collapse
Affiliation(s)
- Sara Costi
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laurel S. Morris
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine A. Kirkwood
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Megan Hoch
- Department of Psychology, University of California Los Angeles, Los Angeles, CA, USA
| | - Morgan Corniquel
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brittany Vo-Le
- Mood and Anxiety Disorders Program, Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA; Michael E. Debakey VA Medical Center, Houston, TX, USA
| | - Tabish Iqbal
- Mood and Anxiety Disorders Program, Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA; Michael E. Debakey VA Medical Center, Houston, TX, USA
| | - Nisha Chadha
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diego A. Pizzagalli
- Department of Psychiatry, Harvard Medical School & McLean Hospital, Belmont, MA, USA
| | - Alexis Whitton
- Department of Psychiatry, Harvard Medical School & McLean Hospital, Belmont, MA, USA,School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Laura Bevilacqua
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manish K. Jha
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stefan Ursu
- Mood and Anxiety Disorders Program, Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA; Michael E. Debakey VA Medical Center, Houston, TX, USA
| | - Alan C Swann
- Mood and Anxiety Disorders Program, Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA; Michael E. Debakey VA Medical Center, Houston, TX, USA
| | - Katherine A. Collins
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Ramiro Salas
- Mood and Anxiety Disorders Program, Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA; Michael E. Debakey VA Medical Center, Houston, TX, USA
| | - Emilia Bagiella
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael K. Parides
- Montefiore Medical Center/Albert Einstein College of Medicine, New York, New York
| | - Emily R. Stern
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA,Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Dan V. Iosifescu
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA,Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Ming-Hu Han
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA,Center for Affective Neuroscience, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sanjay J. Mathew
- Mood and Anxiety Disorders Program, Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, Texas, USA; Michael E. Debakey VA Medical Center, Houston, TX, USA
| | - James W. Murrough
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
21
|
Loiodice S, Drinkenburg WH, Ahnaou A, McCarthy A, Viardot G, Cayre E, Rion B, Bertaina-Anglade V, Mano M, L’Hostis P, Drieu La Rochelle C, Kas MJ, Danjou P. Mismatch negativity as EEG biomarker supporting CNS drug development: a transnosographic and translational study. Transl Psychiatry 2021; 11:253. [PMID: 33927180 PMCID: PMC8085207 DOI: 10.1038/s41398-021-01371-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/25/2021] [Accepted: 04/09/2021] [Indexed: 11/17/2022] Open
Abstract
The lack of translation from basic research into new medicines is a major challenge in CNS drug development. The need to use novel approaches relying on (i) patient clustering based on neurobiology irrespective to symptomatology and (ii) quantitative biomarkers focusing on evolutionarily preserved neurobiological systems allowing back-translation from clinical to nonclinical research has been highlighted. Here we sought to evaluate the mismatch negativity (MMN) response in schizophrenic (SZ) patients, Alzheimer's disease (AD) patients, and age-matched healthy controls. To evaluate back-translation of the MMN response, we developed EEG-based procedures allowing the measurement of MMN-like responses in a rat model of schizophrenia and a mouse model of AD. Our results indicate a significant MMN attenuation in SZ but not in AD patients. Consistently with the clinical findings, we observed a significant attenuation of deviance detection (~104.7%) in rats subchronically exposed to phencyclidine, while no change was observed in APP/PS1 transgenic mice when compared to wild type. This study provides new insight into the cross-disease evaluation of the MMN response. Our findings suggest further investigations to support the identification of neurobehavioral subtypes that may help patients clustering for precision medicine intervention. Furthermore, we provide evidence that MMN could be used as a quantitative/objective efficacy biomarker during both preclinical and clinical stages of SZ drug development.
Collapse
Affiliation(s)
- Simon Loiodice
- Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042, Rennes, France.
| | - Wilhelmus H. Drinkenburg
- grid.419619.20000 0004 0623 0341Department of Neuroscience Discovery, Janssen Research & Development, a Division of Janssen Pharmaceutical NV, Turnhoutseweg 30, B-2340, Beerse, Belgium ,grid.4830.f0000 0004 0407 1981Groningen Institute for Evolutionary Life Sciences, University of Groningen, P.O. Box 11103, 9700 CC, Groningen, The Netherlands
| | - Abdallah Ahnaou
- grid.419619.20000 0004 0623 0341Department of Neuroscience Discovery, Janssen Research & Development, a Division of Janssen Pharmaceutical NV, Turnhoutseweg 30, B-2340, Beerse, Belgium
| | - Andrew McCarthy
- Lilly Research Laboratories, Windlesham, Surrey, GU20 6PH UK
| | - Geoffrey Viardot
- Biotrial Neuroscience, Avenue de Bruxelles, 68350 Didenheim, France
| | - Emilie Cayre
- Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042 Rennes, France
| | - Bertrand Rion
- Biotrial Pharmacology, 7-9 rue Jean-Louis Bertrand, 35042 Rennes, France
| | | | - Marsel Mano
- Biotrial Neuroscience, Avenue de Bruxelles, 68350 Didenheim, France
| | | | | | - Martien J. Kas
- grid.4830.f0000 0004 0407 1981Groningen Institute for Evolutionary Life Sciences, University of Groningen, P.O. Box 11103, 9700 CC, Groningen, The Netherlands
| | - Philippe Danjou
- Biotrial Neuroscience, Avenue de Bruxelles, 68350 Didenheim, France
| |
Collapse
|
22
|
Rocks T, West M, Hockey M, Aslam H, Lane M, Loughman A, Jacka FN, Ruusunen A. Possible use of fermented foods in rehabilitation of anorexia nervosa: the gut microbiota as a modulator. Prog Neuropsychopharmacol Biol Psychiatry 2021; 107:110201. [PMID: 33307114 DOI: 10.1016/j.pnpbp.2020.110201] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 11/23/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022]
Abstract
Anorexia nervosa is a serious psychiatric disorder with high morbidity and mortality rate. Evidence for the optimal psychopharmacological approach to managing the disorder remains limited, with nutritional treatment, focused on weight restoration through the consumption of high energy diet, regarded as one of the fundamental steps in treatment. The human gut microbiome is increasingly recognised for its proposed role in gastrointestinal, metabolic, immune and mental health, all of which may be compromised in individuals with anorexia nervosa. Dietary intake plays an important role in shaping gut microbiota composition, whilst the use of fermented foods, foods with potential psychobiotic properties that deliver live bacteria, bacterial metabolites, prebiotics and energy, have been discussed to a lesser extent. However, fermented foods are of increasing interest due to their potential capacity to affect gut microbiota composition, provide beneficial bacterial metabolites, and confer beneficial outcomes to host health. This review provides an overview of the role of the gut microbiota in relation to the disease pathology in anorexia nervosa and especially focuses on the therapeutic potential of fermented foods, proposed here as a recommended addition to the current nutritional treatment protocols warranting further investigation.
Collapse
Affiliation(s)
- Tetyana Rocks
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia.
| | - Madeline West
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Meghan Hockey
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Hajara Aslam
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Melissa Lane
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Amy Loughman
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia
| | - Felice N Jacka
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia; Centre for Adolescent Health, Murdoch Children's Research Institute, VIC, Australia; Black Dog Institute, NSW, Australia; James Cook University, QLD; Australia
| | - Anu Ruusunen
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, Australia; Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Psychiatry, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
23
|
De Deurwaerdère P, Chagraoui A, Di Giovanni G. Serotonin/dopamine interaction: Electrophysiological and neurochemical evidence. PROGRESS IN BRAIN RESEARCH 2021; 261:161-264. [PMID: 33785130 DOI: 10.1016/bs.pbr.2021.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The interaction between serotonin (5-HT) and dopamine (DA) in the central nervous system (CNS) plays an important role in the adaptive properties of living animals to their environment. These are two modulatory, divergent systems shaping and regulating in a widespread manner the activity of neurobiological networks and their interaction. The concept of one interaction linking these two systems is rather elusive when looking at the mechanisms triggered by these two systems across the CNS. The great variety of their interacting mechanisms is in part due to the diversity of their neuronal origin, the density of their fibers in a given CNS region, the distinct expression of their numerous receptors in the CNS, the heterogeneity of their intracellular signaling pathway that depend on the cellular type expressing their receptors, and the state of activity of neurobiological networks, conditioning the outcome of their mutual influences. Thus, originally conceptualized as inhibition of 5-HT on DA neuron activity and DA neurotransmission, this interaction is nowadays considered as a multifaceted, mutual influence of these two systems in the regulation of CNS functions. These new ways of understanding this interaction are of utmost importance to envision the consequences of their dysfunctions underlined in several CNS diseases. It is also essential to conceive the mechanism of action of psychotropic drugs directly acting on their function including antipsychotic, antidepressant, antiparkinsonian, and drug of abuse together with the development of therapeutic strategies of Alzheimer's diseases, epilepsy, obsessional compulsive disorders. The 5-HT/DA interaction has a long history from the serendipitous discovery of antidepressants and antipsychotics to the future, rationalized treatments of CNS disorders.
Collapse
Affiliation(s)
- Philippe De Deurwaerdère
- Centre National de la Recherche Scientifique, Institut des Neurosciences Intégratives et Cognitives d'Aquitaine, UMR 5287, Bordeaux, France.
| | - Abdeslam Chagraoui
- Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine of Normandy (IRIB), Normandie University, UNIROUEN, INSERM U1239, Rouen, France; Department of Medical Biochemistry, Rouen University Hospital, Rouen, France
| | - Giuseppe Di Giovanni
- Laboratory of Neurophysiology, Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta; Neuroscience Division, School of Biosciences, Cardiff University, Cardiff, United Kingdom.
| |
Collapse
|
24
|
Zwanzger P, Singewald N, Bandelow B. [Pharmacotherapy of anxiety disorders-Guideline-conform treatment and new developments]. DER NERVENARZT 2021; 92:433-440. [PMID: 33502576 DOI: 10.1007/s00115-020-01051-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/11/2020] [Indexed: 01/07/2023]
Abstract
Besides cognitive behavioral therapy (CBT), psychopharmacotherapy belongs to the first-line treatment approaches for anxiety disorders according to all national and international guidelines. According to studies and meta-analyses, modern antidepressants in particular have been proven to be effective. Depending on the substance, there are approvals for panic disorder, generalized anxiety disorder and social phobia. There are also approvals for other substance groups, e.g. anticonvulsants for generalized anxiety disorder. Benzodiazepines should be used with caution in view of the risk of dependency. Although effective and well-tolerated medications are available, up to 30% of patients still do not respond or do not respond adequately to treatment. Consequently, research efforts to develop new substances are important. Based on a better understanding of the complex neurobiological mechanisms underlying anxiety disorders, a large number of substances are currently undergoing clinical trials. Modulators of current and new transmitter systems, in particular the glutamatergic and the endocannabinoid systems as well as neuropeptides, are being discussed as innovative substances. Strategies are also being investigated which, in combination with psychotherapy, aim at optimizing fear extinction memory. First studies are also underway on the use of psychedelic agents in combination with psychotherapy for anxiety.
Collapse
Affiliation(s)
- P Zwanzger
- Fachbereich Psychosomatische Medizin, Kompetenzschwerpunkt Angst, kbo-Inn-Salzach-Klinikum, Gabersee 7, 83512, Wasserburg am Inn, Deutschland. .,Klinik für Psychiatrie und Psychotherapie, Ludwig-Maximilians-Universität München, München, Deutschland.
| | - N Singewald
- Center for Molecular Biosciences Innsbruck (CMBI), Department of Pharmacology and Toxicology, Institute of Pharmacy, Leopold-Franzens-Universität Innsbruck, Innsbruck, Österreich
| | - B Bandelow
- Klinik für Psychiatrie und Psychotherapie, Universitätsmedizin Göttingen, Göttingen, Deutschland
| |
Collapse
|
25
|
Thomson DM, Openshaw RL, Mitchell EJ, Kouskou M, Millan MJ, Mannoury la Cour C, Morris BJ, Pratt JA. Impaired working memory, cognitive flexibility and reward processing in mice genetically lacking Gpr88: Evidence for a key role for Gpr88 in multiple cortico-striatal-thalamic circuits. GENES BRAIN AND BEHAVIOR 2020; 20:e12710. [PMID: 33078498 DOI: 10.1111/gbb.12710] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 12/22/2022]
Abstract
The GPR88 orphan G protein-coupled receptor is expressed throughout the striatum, being preferentially localised in medium spiny neurons. It is also present in lower densities in frontal cortex and thalamus. Rare mutations in humans suggest a role in cognition and motor function, while common variants are associated with psychosis. Here we evaluate the influence of genetic deletion of GPR88 upon performance in translational tasks interrogating motivation, reward evaluation and cognitive function. In an automated radial arm maze 'N-back' working memory task, Gpr88 KO mice showed impaired correct responding, suggesting a role for GPR88 receptors in working memory circuitry. Associative learning performance was similar to wild-type controls in a touchscreen task but performance was impaired at the reversal learning stage, suggesting cognitive inflexibility. Gpr88 KO mice showed higher breakpoints, reduced latencies and lengthened session time in a progressive ratio task consistent with enhanced motivation. Simultaneously, locomotor hyperactivity was apparent in this task, supporting previous findings of actions of GPR88 in a cortico-striatal-thalamic motor loop. Evidence for a role of GPR88 in reward processing was demonstrated in a touchscreen-based equivalent of the Iowa gambling task. Although both Gpr88 KO and wild-type mice showed a preference for an optimum contingency choice, Gpr88 KO mice selected more risky choices at the expense of more advantageous lower risk options. Together these novel data suggest that striatal GPR88 receptors influence activity in a range of procedures integrated by prefrontal, orbitofrontal and anterior cingulate cortico-striatal-thalamic loops leading to altered cognitive, motivational and reward evaluation processes.
Collapse
Affiliation(s)
- David M Thomson
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, G4 0RE, United Kingdom
| | - Rebecca L Openshaw
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, G4 0RE, United Kingdom
| | - Emma J Mitchell
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, G4 0RE, United Kingdom
| | - Marianna Kouskou
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, G4 0RE, United Kingdom
| | - Mark J Millan
- Centre for Therapeutic Innovation-CNS, Institute de Recherche Servier, Croissy-sur-Seine, France
| | | | - Brian J Morris
- Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Judith A Pratt
- Strathclyde Institute of Pharmacy and Biomedical Science, University of Strathclyde, Glasgow, G4 0RE, United Kingdom
| |
Collapse
|
26
|
Ågren R, Zeberg H, Stępniewski TM, Free RB, Reilly SW, Luedtke RR, Århem P, Ciruela F, Sibley DR, Mach RH, Selent J, Nilsson J, Sahlholm K. Ligand with Two Modes of Interaction with the Dopamine D 2 Receptor-An Induced-Fit Mechanism of Insurmountable Antagonism. ACS Chem Neurosci 2020; 11:3130-3143. [PMID: 32865974 PMCID: PMC7553383 DOI: 10.1021/acschemneuro.0c00477] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
A solid understanding of the mechanisms governing ligand binding is crucial for rational design of therapeutics targeting the dopamine D2 receptor (D2R). Here, we use G protein-coupled inward rectifier potassium (GIRK) channel activation in Xenopus oocytes to measure the kinetics of D2R antagonism by a series of aripiprazole analogues, as well as the recovery of dopamine (DA) responsivity upon washout. The aripiprazole analogues comprise an orthosteric and a secondary pharmacophore and differ by the length of the saturated carbon linker joining these two pharmacophores. Two compounds containing 3- and 5-carbon linkers allowed for a similar extent of recovery from antagonism in the presence of 1 or 100 μM DA (>25 and >90% of control, respectively), whereas recovery was less prominent (∼20%) upon washout of the 4-carbon linker compound, SV-III-130, both with 1 and 100 μM DA. Prolonging the coincubation time with SV-III-130 further diminished recovery. Curve-shift experiments were consistent with competition between SV-III-130 and DA. Two mutations in the secondary binding pocket (V91A and E95A) of D2R decreased antagonistic potency and increased recovery from SV-III-130 antagonism, whereas a third mutation (L94A) only increased recovery. Our results suggest that the secondary binding pocket influences recovery from inhibition by the studied aripiprazole analogues. We propose a mechanism, supported by in silico modeling, whereby SV-III-130 initially binds reversibly to the D2R, after which the drug-receptor complex undergoes a slow transition to a second ligand-bound state, which is dependent on secondary binding pocket integrity and irreversible during the time frame of our experiments.
Collapse
Affiliation(s)
- Richard Ågren
- Department
of Clinical Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
- Department
of Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Hugo Zeberg
- Department
of Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Tomasz Maciej Stępniewski
- Research
Programme on Biomedical Informatics (GRIB), Department of Experimental
and Health Sciences of Pompeu Fabra University (UPF)-Hospital del
Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
- InterAx
Biotech AG, PARK innovAARE, 5234 Villigen, Switzerland
- Faculty
of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw 02-089, Poland
| | - R. Benjamin Free
- Molecular
Neuropharmacology Section, National Institute of Neurological Disorders
and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland 20892-3723, United States
| | - Sean W. Reilly
- Department
of Radiology, Division of Nuclear Medicine and Clinical Molecular
Imaging, University of Pennsylvania Perelman
School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Robert R. Luedtke
- Department
of Pharmacology and Neuroscience, University
of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, Texas 76107, United States
| | - Peter Århem
- Department
of Clinical Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
- Department
of Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Francisco Ciruela
- Pharmacology
Unit, Department of Pathology and Experimental Therapeutics, Faculty
of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L’Hospitalet de Llobregat 08907, Spain
- Neuropharmacology
and Pain Group, Neuroscience Program, Institut
d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat 08907, Spain
| | - David R. Sibley
- Molecular
Neuropharmacology Section, National Institute of Neurological Disorders
and Stroke, Intramural Research Program, National Institutes of Health, Bethesda, Maryland 20892-3723, United States
| | - Robert H. Mach
- Department
of Radiology, Division of Nuclear Medicine and Clinical Molecular
Imaging, University of Pennsylvania Perelman
School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Jana Selent
- Research
Programme on Biomedical Informatics (GRIB), Department of Experimental
and Health Sciences of Pompeu Fabra University (UPF)-Hospital del
Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain
| | - Johanna Nilsson
- Department
of Clinical Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Kristoffer Sahlholm
- Department
of Neuroscience, Karolinska Institutet, Stockholm 171 77, Sweden
- Department
of Integrative Medical Biology, Umeå
University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine, Umeå
University, Umeå 901 87, Sweden
| |
Collapse
|
27
|
5-Hydroxytryptophan as adjuvant therapy in treatment of moderate to severe obsessive-compulsive disorder: a double-blind randomized trial with placebo control. Int Clin Psychopharmacol 2020; 35:254-262. [PMID: 32541380 DOI: 10.1097/yic.0000000000000321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
On the basis of numerous previous studies, the serotonergic system plays a role in the pathogenesis of obsessive-compulsive disorder (OCD) and effective agents in this pathway, such as 5-hydroxytryptophan, can potentially contribute to treatment of patients with this disorder. Evaluating the efficacy of 5-hydroxytryptophan in treating OCD was the aim of the present randomized, double-blind, placebo-controlled 12-week trial. In a 12-week, randomized double-blind study, 60 patients with a Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition diagnosis of moderate to severe OCD and a Yale-Brown Obsessive Compulsive Scale (Y-BOCS) score of >21 were randomly assigned to receive either fluoxetine plus placebo or fluoxetine plus 5-hydroxytryptophan (100 mg twice daily). All patients, regardless of their treatment group, received fluoxetine at 20 mg/day for the initial 4 weeks of the study followed by 60 mg/day of fluoxetine for the rest of the trial course. Symptoms were assessed using the Y-BOCS at baseline and weeks 4, 8 and 12. General linear model repeated measure showed significant effects for time × treatment interaction on total Y-BOCS (F = 12.07, df = 2.29, P-value <0.001), obsession (F = 8.25, df = 1.91, P-value = 0.001) and compulsion subscale scores (F = 6.64, df = 2.01, P-value = 0.002). 5-Hydroxytryptophan augmentation therapy demonstrated higher partial and complete treatment response rate (P = 0.032 and P = 0.001, respectively) according to the Y-BOCS total scores. The results of this study confirm that 5-hydroxytryptophan may be effective as an augmentative agent in treatment of moderate-to-severe OCD.
Collapse
|
28
|
El Saftawy EA, Amin NM, Sabry RM, El-Anwar N, Shash RY, Elsebaie EH, Wassef RM. Can Toxoplasma gondii Pave the Road for Dementia? J Parasitol Res 2020; 2020:8859857. [PMID: 32802484 PMCID: PMC7414348 DOI: 10.1155/2020/8859857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/25/2020] [Accepted: 07/03/2020] [Indexed: 01/04/2023] Open
Abstract
Dementia is an ominous neurological disease. Scientists proposed a link between its occurrence and the presence of Toxoplasma gondii (T. gondii). The long-term sequels of anti-Toxoplasma premunition, chiefly dominated by TNF-α, on the neurons and their receptors as the insulin-like growth factor-1 receptor (IGF-1R), which is tangled in cognition and synaptic plasticity, are still not clear. IGF-1R mediates its action via IGF-1, and its depletion is incorporated in the pathogenesis of dementia. The activated TNF-α signaling pathway induces NF-κβ that may induce or inhibit neurogenesis. This study speculates the potential impact of anti-Toxoplasma immune response on the expression of IGF-1R in chronic cerebral toxoplasmosis. The distributive pattern of T. gondii cysts was studied in association with TNF-α serum levels, the in situ expression of NF-κβ, and IGF-1R in mice using the low virulent ME-49 T. gondii strain. There was an elevation of the TNF-α serum level (p value ≤ 0.004) and significant upsurge in NF-κβ whereas IGF-1R was of low abundance (p value < 0.05) compared to the controls. TNF-α had a strong positive correlation with the intracerebral expression of NF-κβ (r value ≈ 0.943, p value ≈ 0.005) and a strong negative correlation to IGF-1R (r value -0.584 and -0.725 for area% and O.D., respectively). This activated TNF-α/NF-κβ keeps T. gondii under control at the expense of IGF-1R expression, depriving neurons of the effect of IGF-1, the receptor's ligand. We therefore deduce that T. gondii immunopathological reaction may be a road paver for developing dementia.
Collapse
Affiliation(s)
- Enas A. El Saftawy
- Medical Parasitology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
- Armed Forces College of Medicine, Cairo, Egypt
| | - Noha M. Amin
- Medical Parasitology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rania M. Sabry
- Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Noha El-Anwar
- Armed Forces College of Medicine, Cairo, Egypt
- Pathology Department, Faculty of Medicine, Tanta University, Egypt
| | - Rania Y. Shash
- Medical Microbiology and Immunology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Eman H. Elsebaie
- Public Health and Community Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rita M. Wassef
- Medical Parasitology Department, Faculty of Medicine, Helwan University, Cairo, Egypt
| |
Collapse
|
29
|
Silote GP, de Oliveira SFS, Ribeiro DE, Machado MS, Andreatini R, Joca SRL, Beijamini V. Ketamine effects on anxiety and fear-related behaviors: Current literature evidence and new findings. Prog Neuropsychopharmacol Biol Psychiatry 2020; 100:109878. [PMID: 31982463 DOI: 10.1016/j.pnpbp.2020.109878] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/16/2020] [Accepted: 01/23/2020] [Indexed: 12/19/2022]
Abstract
Ketamine, a non-competitive N-methyl-D-aspartate (NMDA) receptor antagonist, presents a rapid and sustained antidepressant effect in clinical and preclinical studies. Regarding ketamine effects on anxiety, there is a widespread discordance among pre-clinical studies. To address this issue, the present study reviewed the literature (electronic database MEDLINE) to summarize the profile of ketamine effects in animal tests of anxiety/fear. We found that ketamine anxiety/fear-related effects may depend on the anxiety paradigm, schedule of ketamine administration and tested species. Moreover, there was no report of ketamine effects in animal tests of fear related to panic disorder (PD). Based on that finding, we evaluated if treatment with ketamine and another NMDA antagonist, MK-801, would induce acute and sustained (24 hours later) anxiolytic and/or panicolytic-like effects in animals exposed to the elevated T-maze (ETM). The ETM evaluates, in the same animal, conflict-evoked and fear behaviors, which are related, respectively, to generalized anxiety disorder and PD. Male Wistar rats were systemically treated with racemic ketamine (10, 30 and 80 mg/kg) or MK-801 (0.05 and 0.1 mg/kg) and tested in the ETM in the same day or 24 hours after their administration. Ketamine did not affect the behavioral tasks performed in the ETM acutely or 24 h later. MK-801 impaired inhibitory avoidance in the ETM only at 45 min post-injection, suggesting a rapid but not sustained anxiolytic-like effect. Altogether our results suggest that ketamine might have mixed effects in anxiety tests while it does not affect panic-related behaviors.
Collapse
Affiliation(s)
- Gabriela P Silote
- Biochemistry and Pharmacology Graduate Program, Federal University of Espirito Santo, Vitoria, ES, Brazil; Department of Biomolecular Sciences, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil; Translational Neuropsychiatry Unit (TNU), Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sabrina F S de Oliveira
- Department of Pharmaceutical Sciences, Health Science Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Deidiane E Ribeiro
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Mayara S Machado
- Department of Pharmaceutical Sciences, Health Science Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Roberto Andreatini
- Department of Pharmacology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Sâmia R L Joca
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil; Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Denmark
| | - Vanessa Beijamini
- Biochemistry and Pharmacology Graduate Program, Federal University of Espirito Santo, Vitoria, ES, Brazil; Department of Pharmaceutical Sciences, Health Science Center, Federal University of Espirito Santo, Vitoria, ES, Brazil; Pharmaceutical Sciences Graduate Program, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| |
Collapse
|
30
|
Millan MJ, Dekeyne A, Gobert A, Brocco M, Mannoury la Cour C, Ortuno JC, Watson D, Fone KCF. Dual-acting agents for improving cognition and real-world function in Alzheimer's disease: Focus on 5-HT6 and D3 receptors as hubs. Neuropharmacology 2020; 177:108099. [PMID: 32525060 DOI: 10.1016/j.neuropharm.2020.108099] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 01/01/2023]
Abstract
To date, there are no interventions that impede the inexorable progression of Alzheimer's disease (AD), and currently-available drugs cholinesterase (AChE) inhibitors and the N-Methyl-d-Aspartate receptor antagonist, memantine, offer only modest symptomatic benefit. Moreover, a range of mechanistically-diverse agents (glutamatergic, histaminergic, monoaminergic, cholinergic) have disappointed in clinical trials, alone and/or in association with AChE inhibitors. This includes serotonin (5-HT) receptor-6 antagonists, despite compelling preclinical observations in rodents and primates suggesting a positive influence on cognition. The emphasis has so far been on high selectivity. However, for a multi-factorial disorder like idiopathic AD, 5-HT6 antagonists possessing additional pharmacological actions might be more effective, by analogy to "multi-target" antipsychotics. Based on this notion, drug discovery programmes have coupled 5-HT6 blockade to 5-HT4 agonism and inhibition of AchE. Further, combined 5-HT6/dopamine D3 receptor (D3) antagonists are of especial interest since D3 blockade mirrors 5-HT6 antagonism in exerting broad-based pro-cognitive properties in animals. Moreover, 5-HT6 and dopamine D3 antagonists promote neurocognition and social cognition via both distinctive and convergent actions expressed mainly in frontal cortex, including suppression of mTOR over-activation and reinforcement of cholinergic and glutamatergic transmission. In addition, 5-HT6 blockade affords potential anti-anxiety, anti-depressive and anti-epileptic properties, and antagonising 5-HT6 receptors may be associated with neuroprotective ("disease-modifying") properties. Finally D3 antagonism may counter psychotic episodes and D3 receptors themselves offer a promising hub for multi-target agents. The present article reviews the status of "R and D" into multi-target 5-HT6 and D3 ligands for improved treatment of AD and other neurodegenerative disorders of aging. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.
Collapse
Affiliation(s)
- Mark J Millan
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France.
| | - Anne Dekeyne
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Alain Gobert
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Mauricette Brocco
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Clotilde Mannoury la Cour
- Centre for Therapeutic Innovation in Neuropsychiatry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - Jean-Claude Ortuno
- Centre for Excellence in Chemistry, Institut de Recherche Servier, 78290, Croissy sur Seine, France
| | - David Watson
- School of Life Sciences, Queen's Medical Centre, The University of Nottingham, NG7 2UH, England, UK
| | - Kevin C F Fone
- School of Life Sciences, Queen's Medical Centre, The University of Nottingham, NG7 2UH, England, UK
| |
Collapse
|
31
|
Tan A, Costi S, Morris LS, Van Dam NT, Kautz M, Whitton AE, Friedman AK, Collins KA, Ahle G, Chadha N, Do B, Pizzagalli DA, Iosifescu DV, Nestler EJ, Han MH, Murrough JW. Effects of the KCNQ channel opener ezogabine on functional connectivity of the ventral striatum and clinical symptoms in patients with major depressive disorder. Mol Psychiatry 2020; 25:1323-1333. [PMID: 30385872 PMCID: PMC6494706 DOI: 10.1038/s41380-018-0283-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/14/2018] [Accepted: 10/03/2018] [Indexed: 12/28/2022]
Abstract
Major depressive disorder (MDD) is a leading cause of disability worldwide, yet current treatment strategies remain limited in their mechanistic diversity. Recent evidence has highlighted a promising novel pharmaceutical target-the KCNQ-type potassium channel-for the treatment of depressive disorders, which may exert a therapeutic effect via functional changes within the brain reward system, including the ventral striatum. The current study assessed the effects of the KCNQ channel opener ezogabine (also known as retigabine) on reward circuitry and clinical symptoms in patients with MDD. Eighteen medication-free individuals with MDD currently in a major depressive episode were enrolled in an open-label study and received ezogabine up to 900 mg/day orally over the course of 10 weeks. Resting-state functional magnetic resonance imaging data were collected at baseline and posttreatment to examine brain reward circuitry. Reward learning was measured using a computerized probabilistic reward task. After treatment with ezogabine, subjects exhibited a significant reduction of depressive symptoms (Montgomery-Asberg Depression Rating Scale score change: -13.7 ± 9.7, p < 0.001, d = 2.08) and anhedonic symptoms (Snaith-Hamilton Pleasure Scale score change: -6.1 ± 5.3, p < 0.001, d = 1.00), which remained significant even after controlling for overall depression severity. Improvement in depression was associated with decreased functional connectivity between the ventral caudate and clusters within the mid-cingulate cortex and posterior cingulate cortex (n = 14, voxel-wise p < 0.005). In addition, a subgroup of patients tested with a probabilistic reward task (n = 9) showed increased reward learning following treatment. These findings highlight the KCNQ-type potassium channel as a promising target for future drug discovery efforts in mood disorders.
Collapse
Affiliation(s)
- Aaron Tan
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sara Costi
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laurel S. Morris
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicholas T. Van Dam
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Melbourne School of Psychological Sciences, University of Melbourne, Melbourne, Australia
| | - Marin Kautz
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Allyson K. Friedman
- Department of Biological Sciences, Hunter College, The City University of New York, New York, NY, USA
| | - Katherine A. Collins
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriella Ahle
- Department of Psychology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nisha Chadha
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian Do
- Roski Eye Institute, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | | | - Dan V. Iosifescu
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA,Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| | - Eric J. Nestler
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ming-Hu Han
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James W. Murrough
- Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA,To Whom Correspondence Should Be Addressed: James Murrough, M.D., Ph.D., Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY 10029, USA, Ph: (212) 241-7574, Fax: (212) 241-3354,
| |
Collapse
|
32
|
Rich MC, Sherwood J, Bartley AF, Whitsitt QA, Lee M, Willoughby WR, Dobrunz LE, Bao Y, Lubin FD, Bolding M. Focused ultrasound blood brain barrier opening mediated delivery of MRI-visible albumin nanoclusters to the rat brain for localized drug delivery with temporal control. J Control Release 2020; 324:172-180. [PMID: 32376461 DOI: 10.1016/j.jconrel.2020.04.054] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 12/11/2022]
Abstract
There is an ongoing need for noninvasive tools to manipulate brain activity with molecular, spatial and temporal specificity. Here we have investigated the use of MRI-visible, albumin-based nanoclusters for noninvasive, localized and temporally specific drug delivery to the rat brain. We demonstrated that IV injected nanoclusters could be deposited into target brain regions via focused ultrasound facilitated blood brain barrier opening. We showed that nanocluster location could be confirmed in vivo with MRI. Additionally, following confirmation of nanocluster delivery, release of the nanocluster payload into brain tissue can be triggered by a second focused ultrasound treatment performed without circulating microbubbles. Release of glutamate from nanoclusters in vivo caused enhanced c-Fos expression, indicating that the loading capacity of the nanoclusters is sufficient to induce neuronal activation. This novel technique for noninvasive stereotactic drug delivery to the brain with temporal specificity could provide a new way to study brain circuits in vivo preclinically with high relevance for clinical translation.
Collapse
Affiliation(s)
- Megan C Rich
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Jennifer Sherwood
- Department of Chemical and Biological Engineering, University of Alabama at Tuscaloosa, Tuscaloosa, AL 35487, USA
| | - Aundrea F Bartley
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Quentin A Whitsitt
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Magdelene Lee
- Department of Chemical and Biological Engineering, University of Alabama at Tuscaloosa, Tuscaloosa, AL 35487, USA
| | - W R Willoughby
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Lynn E Dobrunz
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Yuping Bao
- Department of Chemical and Biological Engineering, University of Alabama at Tuscaloosa, Tuscaloosa, AL 35487, USA.
| | - Farah D Lubin
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Mark Bolding
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
33
|
Harro J, Aadamsoo K, Rootslane L, Laius O, O'Leary A, Adomaitiene V, Kupca B, Lehtmets A, Navickas A, Rancans E, Taube M, Terauds E, Pops K. Comparison of psychotropic medication use in the Baltic countries. Nord J Psychiatry 2020; 74:301-306. [PMID: 31889460 DOI: 10.1080/08039488.2019.1707283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Purpose: While the pivotal role of pharmacotherapy in psychiatry is universal, significant regional differences exist in drug use patterns. Herewith we compare the use of ATC psychotropic drugs (N05, psycholeptics and N06A, antidepressants) in 2010-2015 in the three Baltic Countries with reference to the Nordic Countries.Methods: Data were obtained from the national authorities on medicines as expressed in DDD per 1000 inhabitants per day. A semi-structured questionnaire was used for expert statements on the rationale of current use of medicines.Results: During the observation period the use of antipsychotics, anxiolytics, hypnotics and sedatives, and antidepressants steadily increased, while the growth in use of anxiolytics stagnated in the more recent years. Antipsychotic use was the largest in Lithuania and the lowest in Estonia. The use on anxiolytics in Lithuania was more than twice of that in Estonia and Latvia. Conversely, the use of hypnotics and sedatives was about three times higher in Estonia than in Latvia or Lithuania. Antidepressant use was dominated by the selective serotonin reuptake inhibitors in all three countries, but overall was much lower in Latvia as compared to Lithuania and Estonia. As compared to the Nordic Countries in 2015, antidepressants are used at much lower level throughout Baltics, probably reflecting underdiagnostics of depression and anxiety disorders.Conclusion: While the health-care expenditures in Estonia, Latvia and Lithuania are largely similar, as is the cultural and recent political background of these EU member countries, the extent and the pattern of psychotropic drug use is remarkably variable.
Collapse
Affiliation(s)
- Jaanus Harro
- North Estonia Medical Centre, Psychiatry Clinic, Tallinn, Estonia.,Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Tartu, Estonia
| | - Kaire Aadamsoo
- North Estonia Medical Centre, Psychiatry Clinic, Tallinn, Estonia
| | - Ly Rootslane
- Estonian State Agency of Medicines, Tartu, Estonia
| | - Ott Laius
- Estonian State Agency of Medicines, Tartu, Estonia.,Department of Traumatology and Orthopedics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Aet O'Leary
- Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Tartu, Estonia.,Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Laboratory of Translational Psychiatry, University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Biruta Kupca
- Department of Psychiatry and Narcology, Riga Stradins University, Riga, Latvia
| | | | - Alvydas Navickas
- Clinic of Psychiatry, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Elmars Rancans
- Department of Psychiatry and Narcology, Riga Stradins University, Riga, Latvia
| | - Maris Taube
- Department of Psychiatry and Narcology, Riga Stradins University, Riga, Latvia
| | - Elmars Terauds
- Department of Psychiatry and Narcology, Riga Stradins University, Riga, Latvia
| | | |
Collapse
|
34
|
Riesel A. The erring brain: Error-related negativity as an endophenotype for OCD-A review and meta-analysis. Psychophysiology 2020; 56:e13348. [PMID: 30838682 DOI: 10.1111/psyp.13348] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/12/2018] [Accepted: 01/20/2019] [Indexed: 12/29/2022]
Abstract
Obsessive-compulsive disorder (OCD) is a complex and heterogeneous disorder that is associated with high personal and societal costs. Feelings of doubt, worry, and repetitive behavior, key symptoms of OCD, have been linked to hyperactive error signals in the brain. The error-related negativity (ERN) represents a validated marker of error processing in the ERP. Increased ERN amplitudes in OCD have been reported very robustly over the last 20 years. This article integrates results from 38 studies analyzing the ERN in OCD, using a quantitative meta-analysis. Meta-regressions were used to examine potential moderators such as task type, symptom severity, age, and sample size. The meta-analysis reveals a robust increase of ERN in OCD patients compared to healthy participants in response-conflict tasks (SMD -0.55) that is not modulated by symptom severity and age. No increase in ERN in OCD was observed in tasks that do not induce response conflict (SMD -0.10). In addition to the meta-analysis, the current article reviews evidence supporting that increased ERN amplitudes in OCD fulfill central criteria for an endophenotype. Further, the specificity of increased ERN amplitudes for OCD and its suitability as a potential transdiagnostic endophenotype is discussed. Finally, the clinical utility and clinical applications are examined. Overall, the evidence that increased ERN amplitudes represent a promising endophenotype indicating vulnerability for OCD is compelling. Furthermore, alterations in ERN are not limited to OCD and may constitute a transdiagnostic endophenotype. Altered neural error signals might serve as a diagnostic or predictive marker and represent a promising target for interventions.
Collapse
Affiliation(s)
- Anja Riesel
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
35
|
Mok SWF, Wong VKW, Lo HH, de Seabra Rodrigues Dias IR, Leung ELH, Law BYK, Liu L. Natural products-based polypharmacological modulation of the peripheral immune system for the treatment of neuropsychiatric disorders. Pharmacol Ther 2020; 208:107480. [DOI: 10.1016/j.pharmthera.2020.107480] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 12/31/2019] [Indexed: 02/06/2023]
|
36
|
Translational Studies in the Complex Role of Neurotransmitter Systems in Anxiety and Anxiety Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1191:121-140. [PMID: 32002926 DOI: 10.1007/978-981-32-9705-0_8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Discovery of innovative anxiolytics is severely hampering. Existing anxiolytics are developed decades ago and are still the therapeutics of choice. Moreover, lack of new drug targets forecasts a severe jeopardy in the future treatment of the huge population of CNS-diseased patients. We simply lack the knowledge on what is wrong in brains of anxious people (normal and diseased). Translational research, based on interacting clinical and preclinical research, is extremely urgent. In this endeavor, genetic and genomic approaches are part of the spectrum of contributing factors. We focus on three druggable targets: serotonin transporter, 5-HT1A, and GABAA receptors. It is still uncertain whether and how these targets are involved in normal and diseased anxiety processes. For serotonergic anxiolytics, the slow onset of action points to indirect effects leading to plasticity changes in brain systems leading to reduced anxiety. For GABAA benzodiazepine drugs, acute anxiolytic effects are found indicating primary mechanisms directly influencing anxiety processes. Close translational collaboration between fundamental academic and discovery research will lead to badly needed breakthroughs in the search for new anxiolytics.
Collapse
|
37
|
Vaudano E. Public–private partnerships as enablers of progress in the fight against mental disorders: The example of the European Innovative Medicines Initiative. Eur Psychiatry 2020; 50:57-59. [DOI: 10.1016/j.eurpsy.2017.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/04/2017] [Accepted: 10/04/2017] [Indexed: 10/18/2022] Open
|
38
|
Sinopoli VM, Erdman L, Burton CL, Park LS, Dupuis A, Shan J, Goodale T, Shaheen SM, Crosbie J, Schachar RJ, Arnold PD. Serotonin system genes and obsessive-compulsive trait dimensions in a population-based, pediatric sample: a genetic association study. J Child Psychol Psychiatry 2019; 60:1289-1299. [PMID: 31321769 DOI: 10.1111/jcpp.13079] [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] [Accepted: 05/03/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Serotonin system genes are commonly studied in obsessive-compulsive disorder (OCD), but genetic studies to date have produced inconsistent results, possibly because phenotypic heterogeneity has not been adequately accounted for. In this paper, we studied candidate serotonergic genes and homogenous phenotypic subgroups as presented through obsessive-compulsive (OC) trait dimensions in a general population of children and adolescents. We hypothesized that different serotonergic gene variants are associated with different OC trait dimensions and, furthermore, that they vary by sex. METHODS Obsessive-compulsive trait dimensions (Cleaning/Contamination, Counting/Checking, Symmetry/Ordering, Superstition, Rumination, and Hoarding) were examined in a total of 5,213 pediatric participants in the community using the Toronto Obsessive-Compulsive Scale (TOCS). We genotyped candidate serotonin genes (directly genotyping the 5-HTTLPR polymorphism in SLC6A4 for 2018 individuals and using single nucleotide polymorphism (SNP) array data for genes SLC6A4, HTR2A, and HTR1B for 4711 individuals). We assessed the association between variants across these genes and each of the OC trait dimensions, within males and females separately. We analyzed OC traits as both (a) dichotomized based on a threshold value and (b) quantitative scores. RESULTS The [LG + S] variant in 5-HTTLPR was significantly associated with hoarding in males (p-value of 0.003 and 0.004 for categorical and continuous analyses, respectively). There were no significant findings for 5-HTTLPR in females. Using SNP array data, there were significant findings for rumination in males for HTR2A SNPs (p-value of 1.04e-6 to 5.20e-6). CONCLUSIONS This represents the first genetic association study of OC trait dimensions in a community-based pediatric sample. Our strongest results indicate that hoarding and rumination may be distinct in their association with serotonin gene variants and that serotonin gene variation may be specific to sex. Future genetic association studies in OCD should properly account for heterogeneity, using homogenous subgroups stratified by symptom dimension, sex, and age group.
Collapse
Affiliation(s)
- Vanessa M Sinopoli
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Program in Genetics & Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Lauren Erdman
- Program in Genetics & Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Christie L Burton
- Program in Genetics & Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Laura S Park
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Program in Genetics & Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Annie Dupuis
- Clinical Research Services, The Hospital for Sick Children, Toronto, ON, Canada.,Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Janet Shan
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Tara Goodale
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - S-M Shaheen
- Mathison Centre for Mental Health Research & Education, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Jennifer Crosbie
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Russell J Schachar
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Paul D Arnold
- Program in Genetics & Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Mathison Centre for Mental Health Research & Education, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Departments of Psychiatry and Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
39
|
Kathuria A, Lopez-Lengowski K, Watmuff B, McPhie D, Cohen BM, Karmacharya R. Synaptic deficits in iPSC-derived cortical interneurons in schizophrenia are mediated by NLGN2 and rescued by N-acetylcysteine. Transl Psychiatry 2019; 9:321. [PMID: 31780643 PMCID: PMC6882825 DOI: 10.1038/s41398-019-0660-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/06/2019] [Accepted: 10/20/2019] [Indexed: 02/08/2023] Open
Abstract
Human postmortem studies suggest a major role for abnormalities in GABAergic interneurons in the prefrontal cortex in schizophrenia. Cortical interneurons differentiated from induced pluripotent stem cells (iPSCs) of schizophrenia subjects showed significantly lower levels of glutamate decarboxylase 67 (GAD67), replicating findings from multiple postmortem studies, as well as reduced levels of synaptic proteins gehpyrin and NLGN2. Co-cultures of the interneurons with excitatory cortical pyramidal neurons from schizophrenia iPSCs showed reduced synaptic puncta density and lower action potential frequency. NLGN2 overexpression in schizophrenia neurons rescued synaptic puncta deficits while NLGN2 knockdown in healthy neurons resulted in reduced synaptic puncta density. Schizophrenia interneurons also had significantly smaller nuclear area, suggesting an innate oxidative stressed state. The antioxidant N-acetylcysteine increased the nuclear area in schizophrenia interneurons, increased NLGN2 expression and rescued synaptic deficits. These results implicate specific deficiencies in the synaptic machinery in cortical interneurons as critical regulators of synaptic connections in schizophrenia and point to a nexus between oxidative stress and NLGN2 expression in mediating synaptic deficits in schizophrenia.
Collapse
Affiliation(s)
- Annie Kathuria
- 0000 0004 0386 9924grid.32224.35Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA USA ,grid.66859.34Chemical Biology and Therapeutic Science Program, Broad Institute of MIT & Harvard, Cambridge, MA USA ,000000041936754Xgrid.38142.3cDepartment of Psychiatry, Harvard Medical School, Boston, MA USA
| | - Kara Lopez-Lengowski
- 0000 0004 0386 9924grid.32224.35Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA USA ,grid.66859.34Chemical Biology and Therapeutic Science Program, Broad Institute of MIT & Harvard, Cambridge, MA USA
| | - Bradley Watmuff
- 0000 0004 0386 9924grid.32224.35Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA USA ,grid.66859.34Chemical Biology and Therapeutic Science Program, Broad Institute of MIT & Harvard, Cambridge, MA USA ,000000041936754Xgrid.38142.3cDepartment of Psychiatry, Harvard Medical School, Boston, MA USA
| | - Donna McPhie
- 000000041936754Xgrid.38142.3cDepartment of Psychiatry, Harvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA USA
| | - Bruce M. Cohen
- 000000041936754Xgrid.38142.3cDepartment of Psychiatry, Harvard Medical School, Boston, MA USA ,0000 0000 8795 072Xgrid.240206.2Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA USA
| | - Rakesh Karmacharya
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA. .,Chemical Biology and Therapeutic Science Program, Broad Institute of MIT & Harvard, Cambridge, MA, USA. .,Department of Psychiatry, Harvard Medical School, Boston, MA, USA. .,Schizophrenia and Bipolar Disorder Program, McLean Hospital, Belmont, MA, USA. .,Graduate Program in Chemical Biology, Harvard University, Cambridge, MA, USA. .,Program in Neuroscience, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
40
|
Smith SJ, Sümbül U, Graybuck LT, Collman F, Seshamani S, Gala R, Gliko O, Elabbady L, Miller JA, Bakken TE, Rossier J, Yao Z, Lein E, Zeng H, Tasic B, Hawrylycz M. Single-cell transcriptomic evidence for dense intracortical neuropeptide networks. eLife 2019; 8:47889. [PMID: 31710287 PMCID: PMC6881117 DOI: 10.7554/elife.47889] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 11/10/2019] [Indexed: 12/19/2022] Open
Abstract
Seeking new insights into the homeostasis, modulation and plasticity of cortical synaptic networks, we have analyzed results from a single-cell RNA-seq study of 22,439 mouse neocortical neurons. Our analysis exposes transcriptomic evidence for dozens of molecularly distinct neuropeptidergic modulatory networks that directly interconnect all cortical neurons. This evidence begins with a discovery that transcripts of one or more neuropeptide precursor (NPP) and one or more neuropeptide-selective G-protein-coupled receptor (NP-GPCR) genes are highly abundant in all, or very nearly all, cortical neurons. Individual neurons express diverse subsets of NP signaling genes from palettes encoding 18 NPPs and 29 NP-GPCRs. These 47 genes comprise 37 cognate NPP/NP-GPCR pairs, implying the likelihood of local neuropeptide signaling. Here, we use neuron-type-specific patterns of NP gene expression to offer specific, testable predictions regarding 37 peptidergic neuromodulatory networks that may play prominent roles in cortical homeostasis and plasticity.
Collapse
Affiliation(s)
| | - Uygar Sümbül
- Allen Institute for Brain Science, Seattle, United States
| | | | | | | | - Rohan Gala
- Allen Institute for Brain Science, Seattle, United States
| | - Olga Gliko
- Allen Institute for Brain Science, Seattle, United States
| | - Leila Elabbady
- Allen Institute for Brain Science, Seattle, United States
| | | | | | - Jean Rossier
- Neuroscience Paris Seine, Sorbonne Université, Paris, France
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, United States
| | - Ed Lein
- Allen Institute for Brain Science, Seattle, United States
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, United States
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Seattle, United States
| | | |
Collapse
|
41
|
Sartori SB, Singewald N. Novel pharmacological targets in drug development for the treatment of anxiety and anxiety-related disorders. Pharmacol Ther 2019; 204:107402. [PMID: 31470029 DOI: 10.1016/j.pharmthera.2019.107402] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/19/2019] [Indexed: 12/24/2022]
Abstract
Current medication for anxiety disorders is suboptimal in terms of efficiency and tolerability, highlighting the need for improved drug treatments. In this review an overview of drugs being studied in different phases of clinical trials for their potential in the treatment of fear-, anxiety- and trauma-related disorders is presented. One strategy followed in drug development is refining and improving compounds interacting with existing anxiolytic drug targets, such as serotonergic and prototypical GABAergic benzodiazepines. A more innovative approach involves the search for compounds with novel mechanisms of anxiolytic action using the growing knowledge base concerning the relevant neurocircuitries and neurobiological mechanisms underlying pathological fear and anxiety. The target systems evaluated in clinical trials include glutamate, endocannabinoid and neuropeptide systems, as well as ion channels and targets derived from phytochemicals. Examples of promising novel candidates currently in clinical development for generalised anxiety disorder, social anxiety disorder, panic disorder, obsessive compulsive disorder or post-traumatic stress disorder include ketamine, riluzole, xenon with one common pharmacological action of modulation of glutamatergic neurotransmission, as well as the neurosteroid aloradine. Finally, compounds such as D-cycloserine, MDMA, L-DOPA and cannabinoids have shown efficacy in enhancing fear-extinction learning in humans. They are thus investigated in clinical trials as an augmentative strategy for speeding up and enhancing the long-term effectiveness of exposure-based psychotherapy, which could render chronic anxiolytic drug treatment dispensable for many patients. These efforts are indicative of a rekindled interest and renewed optimism in the anxiety drug discovery field, after decades of relative stagnation.
Collapse
Affiliation(s)
- Simone B Sartori
- Institute of Pharmacy, Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck (CMBI), Leopold Franzens University Innsbruck, Innsbruck, Austria
| | - Nicolas Singewald
- Institute of Pharmacy, Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck (CMBI), Leopold Franzens University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
42
|
Hassani SA, Lendor S, Boyaci E, Pawliszyn J, Womelsdorf T. Multineuromodulator measurements across fronto-striatal network areas of the behaving macaque using solid-phase microextraction. J Neurophysiol 2019; 122:1649-1660. [PMID: 31433731 DOI: 10.1152/jn.00321.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Different neuromodulators rarely act independent from each other to modify neural processes but are instead coreleased, gated, or modulated. To understand this interdependence of neuromodulators and their collective influence on local circuits during different brain states, it is necessary to reliably extract local concentrations of multiple neuromodulators in vivo. Here we describe results using solid-phase microextraction (SPME), a method providing sensitive, multineuromodulator measurements. SPME is a sampling method that is coupled with mass spectrometry to quantify collected analytes. Reliable measurements of glutamate, dopamine, acetylcholine, and choline were made simultaneously within frontal cortex and striatum of two macaque monkeys (Macaca mulatta) during goal-directed behavior. We find glutamate concentrations several orders of magnitude higher than acetylcholine and dopamine in all brain regions. Dopamine was reliably detected in the striatum at tenfold higher concentrations than acetylcholine. Acetylcholine and choline concentrations were detected with high consistency across brain areas within monkeys and between monkeys. These findings illustrate that SPME microprobes provide a versatile novel tool to characterize multiple neuromodulators across different brain areas in vivo to understand the interdependence and covariation of neuromodulators during goal-directed behavior. Such data would be important to better distinguish between different behavioral states and characterize dysfunctional brain states that may be evident in psychiatric disorders.NEW & NOTEWORTHY Our paper reports a reliable and sensitive novel method for measuring the absolute concentrations of glutamate, acetylcholine, choline, dopamine, and serotonin in brain circuits in vivo. We show that this method reliably samples multiple neurochemicals in three brain areas simultaneously while nonhuman primates are engaged in goal-directed behavior. We further describe how the methodology we describe here may be used by electrophysiologists as a low-barrier-to-entry tool for measuring multiple neurochemicals.
Collapse
Affiliation(s)
- Seyed-Alireza Hassani
- Department of Psychology, Vanderbilt University, Nashville, Tennessee.,Department of Biology, Centre for Vision Research, York University, Toronto, Ontario, Canada
| | - Sofia Lendor
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Ezel Boyaci
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Janusz Pawliszyn
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, Canada
| | - Thilo Womelsdorf
- Department of Psychology, Vanderbilt University, Nashville, Tennessee.,Department of Biology, Centre for Vision Research, York University, Toronto, Ontario, Canada
| |
Collapse
|
43
|
Kessing LV, Rytgaard HC, Gerds TA, Berk M, Ekstrøm CT, Andersen PK. New drug candidates for bipolar disorder-A nation-wide population-based study. Bipolar Disord 2019; 21:410-418. [PMID: 30873730 DOI: 10.1111/bdi.12772] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Drug repurposing is an increasingly promising idea in many fields of medicine. We systematically used Danish nation-wide population-based registers to investigate whether continued use of non-aspirin non-steroidal anti-inflammatory drugs (NSAIDs), low-dose aspirin, high-dose aspirin, statins, allopurinol, and angiotensin agents decrease the rate of incident mania/bipolar disorder. METHODS A nation-wide population-based longitudinal study using Poisson regression analyses including all persons in Denmark who purchased the exposure medication of interest and a random sample of 30% of the Danish population. The follow-up period comprised a 10 years period from 2005 to 2015. Two different outcome measures were included, (1) a diagnosis of mania/bipolar disorder at a psychiatric hospital contact as inpatient or outpatient and (2) a combined measure of a diagnosis of mania/bipolar disorder or initiation of lithium use. RESULTS A total of 1,605,365 subjects were exposed to one of the six drugs of interest during the exposure period from 2005 to 2015, median age 57 years [quartiles: 43;69], and female proportion of 53.1%. Continued use of low-dose aspirin, statins, and angiotensin agents were associated with decreased rates of incident mania/bipolar disorder on both outcome measures. Continued uses of non-aspirin NSAIDs as well as high-dose aspirin were associated with an increased rate of incident bipolar disorder. There were no statistically significant associations for allopurinol. CONCLUSIONS The study supports the potential of agents acting on inflammation and the stress response system in bipolar disorder and illustrates that population-based registers can be used to systematically identify drugs with repurposing potentials.
Collapse
Affiliation(s)
- Lars V Kessing
- Psychiatric Center Copenhagen, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Helene C Rytgaard
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Thomas A Gerds
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Michael Berk
- School of Medicine, Deakin University, Melbourne, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, Department of Psychiatry, Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Claus T Ekstrøm
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Per K Andersen
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
44
|
Aitta-Aho T, Maksimovic M, Dahl K, Sprengel R, Korpi ER. Attenuation of Novelty-Induced Hyperactivity of Gria1-/- Mice by Cannabidiol and Hippocampal Inhibitory Chemogenetics. Front Pharmacol 2019; 10:309. [PMID: 30984001 PMCID: PMC6449460 DOI: 10.3389/fphar.2019.00309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/13/2019] [Indexed: 11/13/2022] Open
Abstract
Gene-targeted mice with deficient AMPA receptor GluA1 subunits (Gria1-/- mice) show robust hyperlocomotion in a novel environment, suggesting them to constitute a model for hyperactivity disorders such as mania, schizophrenia and attention deficit hyperactivity disorder. This behavioral alteration has been associated with increased neuronal activation in the hippocampus, and it can be attenuated by chronic treatment with antimanic drugs, such as lithium, valproic acid, and lamotrigine. Now we found that systemic cannabidiol strongly blunted the hyperactivity and the hippocampal c-Fos expression of the Gria1-/- mice, while not affecting the wild-type littermate controls. Acute bilateral intra-dorsal hippocampal infusion of cannabidiol partially blocked the hyperactivity of the Gria1-/- mice, but had no effect on wild-types. The activation of the inhibitory DREADD receptor hM4Gi in the dorsal hippocampus by clozapine-N-oxide robustly inhibited the hyperactivity of the Gria1-/- mice, but had no effect on the locomotion of wild-type mice. Our results show that enhanced neuronal excitability in the hippocampus is associated with pronounced novelty-induced hyperactivity of GluA1 subunit-deficient mice. When this enhanced response of hippocampal neurons to novel stimuli is specifically reduced in the hippocampus by pharmacological treatment or by chemogenetic inhibition, Gria1-/- mice recover from behavioral hyperactivity, suggesting a hippocampal dysfunction in hyperactive behaviors that can be treated with cannabidiol.
Collapse
Affiliation(s)
- Teemu Aitta-Aho
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Milica Maksimovic
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kristiina Dahl
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Rolf Sprengel
- Research Group of the Max Planck Institute for Medical Research at the Institute of Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Esa R Korpi
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
45
|
Scalable Measurements of Intrinsic Excitability in Human iPS Cell-Derived Excitatory Neurons Using All-Optical Electrophysiology. Neurochem Res 2019; 44:714-725. [PMID: 30603979 DOI: 10.1007/s11064-018-2694-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 12/02/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
Induced pluripotent stem (iPS) cells offer the exciting opportunity for modeling neurological disorders in vitro in the context of a human genetic background. While significant progress has been made in advancing the use of iPS cell-based disease models, there remains an unmet need to characterize the electrophysiological profile of individual neurons with sufficient throughput to enable statistically robust assessment of disease phenotypes and pharmacological modulation. Here, we describe the Optopatch platform technology that utilizes optogenetics to both stimulate and record action potentials (APs) from human iPS cell-derived excitatory neurons with similar information content to manual patch clamp electrophysiology, but with ~ 3 orders of magnitude greater throughput. Cortical excitatory neurons were produced using the NGN2 transcriptional programming approach and cultured in the presence of rodent glial cells. Characterization of the neuronal preparations using immunocytochemistry and qRT-PCR assays reveals an enrichment of neuronal and glutamatergic markers as well as select ion channels. We demonstrate the scale of our intrinsic cellular excitability assay using pharmacological assessment with select ion channel modulators quinidine and retigabine, by measuring changes in both spike timing and waveform properties. The Optopatch platform in human iPS cell-derived cortical excitatory neurons has the potential for detailed phenotype and pharmacology evaluation, which can serve as the basis of cellular disease model exploration for drug discovery and phenotypic screening efforts.
Collapse
|
46
|
Mata R, Figueroa M, Navarrete A, Rivero-Cruz I. Chemistry and Biology of Selected Mexican Medicinal Plants. PROGRESS IN THE CHEMISTRY OF ORGANIC NATURAL PRODUCTS 2019; 108:1-142. [PMID: 30924013 DOI: 10.1007/978-3-030-01099-7_1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Herbal medicines are an integral element of alternative medical care in Mexico, and the best testimony to their efficacy and cultural value is their persistence in contemporary Mexican marketplaces where the highest percentages of medicinal and aromatic plants are sold. This chapter summarizes current trends in research on medicinal plants in Mexico, with emphasis on work carried out at the authors' laboratories. The most relevant phytochemical and pharmacological profiles of a selected group of plants used widely for treating major national health problems are described.From this contribution, it is evident that in the last five decades a significant amount of research on medicinal plants has been performed by Mexican scientists. Such efforts have led to the publication of many research papers in noted peer-reviewed journals and technical books. The isolation and structural characterization of hundreds of bioactive secondary metabolites have been accomplished, and most importantly, these studies have tended to support the ethnomedical uses of many different species. A multidisciplinary approach for investigating these plants has led to an increased emphasis on areas such as phytopharmacology, phytotoxicology, quality control, regulation, and conservation issues for these valuable resources. The medicinal plants analyzed so far have shown a very broad chemical diversity of their constituents, which have a high potential for exhibiting novel mechanistic effects biologically. The chapter shows also that there is need to conduct additional clinical studies on herbal drugs, in particular because the longstanding traditional evidence for their safety is not always sufficient to assure their rational use. There is also need to move to "omics" approaches for investigating the holistic effect and the influence of groups of phytochemicals on the whole organism. Mexican scientists may be expected to have bright prospects in this regard, which will imbue medicinal plant research with a new dynamism in the future.
Collapse
Affiliation(s)
- Rachel Mata
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, México.
| | - Mario Figueroa
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, México.
| | - Andrés Navarrete
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Isabel Rivero-Cruz
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
47
|
Kessing LV, Rytgaard HC, Gerds TA, Berk M, Ekstrøm CT, Andersen PK. New drug candidates for depression - a nationwide population-based study. Acta Psychiatr Scand 2019; 139:68-77. [PMID: 30182363 DOI: 10.1111/acps.12957] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/15/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To investigate whether continued use of non-aspirin NSAID, low-dose aspirin, high-dose aspirin, statins, allopurinol and angiotensin agents decreases the rate of incident depression using Danish nationwide population-based registers. METHODS All persons in Denmark who purchased the exposure medications of interest between 1995 and 2015 and a random sample of 30% of the Danish population was included in the study. Two different outcome measures were included, (i) a diagnosis of depressive disorder at a psychiatric hospital as in-patient or out-patient and (ii) a combined measure of a diagnosis of depression or use of antidepressants. RESULTS A total of 1 576 253 subjects were exposed to one of the six drugs of interest during the exposure period from 2005 to 2015. Continued use of low-dose aspirin, statins, allopurinol and angiotensin agents was associated with a decreased rate of incident depression according to both outcome measures. Continued uses of non-aspirin NSAIDs as well as high-dose aspirin were associated with an increased rate of incident depression. CONCLUSION The findings support the potential of agents acting on inflammation and the stress response system in depression as well as the potential of population-based registers to systematically identify drugs with repurposing potential.
Collapse
Affiliation(s)
- L V Kessing
- Copenhagen Affective Disorder reaserch Center (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - H C Rytgaard
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - T A Gerds
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - M Berk
- School of Medicine, Deakin University, Geelong, Vic, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry, and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, Vic, Australia
| | - C T Ekstrøm
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - P K Andersen
- Department of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
48
|
Huang C, Zhong Q, Tang L, Wang H, Xu J, Zhou Z. Discovery of 2‐(3,4‐dialkoxyphenyl)‐2‐(substituted pyridazin‐3‐yl)acetonitriles as phosphodiesterase 4 inhibitors with anti‐neuroinflammation potential based on three‐dimensional quantitative structure–activity relationship study. Chem Biol Drug Des 2018; 93:484-502. [DOI: 10.1111/cbdd.13438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/10/2018] [Accepted: 10/27/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Chang Huang
- Department of Neuropharmacology and Novel Drug DiscoverySchool of Pharmaceutical SciencesSouthern Medical University Guangzhou China
- Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical University Guangzhou China
| | - Qiu‐Ping Zhong
- Department of Neuropharmacology and Novel Drug DiscoverySchool of Pharmaceutical SciencesSouthern Medical University Guangzhou China
- Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical University Guangzhou China
| | - Lv Tang
- Department of Neuropharmacology and Novel Drug DiscoverySchool of Pharmaceutical SciencesSouthern Medical University Guangzhou China
- Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical University Guangzhou China
| | - Hai‐Tao Wang
- Department of Neuropharmacology and Novel Drug DiscoverySchool of Pharmaceutical SciencesSouthern Medical University Guangzhou China
- Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical University Guangzhou China
| | - Jiang‐Ping Xu
- Department of Neuropharmacology and Novel Drug DiscoverySchool of Pharmaceutical SciencesSouthern Medical University Guangzhou China
- Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical University Guangzhou China
| | - Zhong‐Zhen Zhou
- Department of Neuropharmacology and Novel Drug DiscoverySchool of Pharmaceutical SciencesSouthern Medical University Guangzhou China
- Guangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical University Guangzhou China
| |
Collapse
|
49
|
Hökfelt T, Barde S, Xu ZQD, Kuteeva E, Rüegg J, Le Maitre E, Risling M, Kehr J, Ihnatko R, Theodorsson E, Palkovits M, Deakin W, Bagdy G, Juhasz G, Prud’homme HJ, Mechawar N, Diaz-Heijtz R, Ögren SO. Neuropeptide and Small Transmitter Coexistence: Fundamental Studies and Relevance to Mental Illness. Front Neural Circuits 2018; 12:106. [PMID: 30627087 PMCID: PMC6309708 DOI: 10.3389/fncir.2018.00106] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/05/2018] [Indexed: 12/31/2022] Open
Abstract
Neuropeptides are auxiliary messenger molecules that always co-exist in nerve cells with one or more small molecule (classic) neurotransmitters. Neuropeptides act both as transmitters and trophic factors, and play a role particularly when the nervous system is challenged, as by injury, pain or stress. Here neuropeptides and coexistence in mammals are reviewed, but with special focus on the 29/30 amino acid galanin and its three receptors GalR1, -R2 and -R3. In particular, galanin's role as a co-transmitter in both rodent and human noradrenergic locus coeruleus (LC) neurons is addressed. Extensive experimental animal data strongly suggest a role for the galanin system in depression-like behavior. The translational potential of these results was tested by studying the galanin system in postmortem human brains, first in normal brains, and then in a comparison of five regions of brains obtained from depressed people who committed suicide, and from matched controls. The distribution of galanin and the four galanin system transcripts in the normal human brain was determined, and selective and parallel changes in levels of transcripts and DNA methylation for galanin and its three receptors were assessed in depressed patients who committed suicide: upregulation of transcripts, e.g., for galanin and GalR3 in LC, paralleled by a decrease in DNA methylation, suggesting involvement of epigenetic mechanisms. It is hypothesized that, when exposed to severe stress, the noradrenergic LC neurons fire in bursts and release galanin from their soma/dendrites. Galanin then acts on somato-dendritic, inhibitory galanin autoreceptors, opening potassium channels and inhibiting firing. The purpose of these autoreceptors is to act as a 'brake' to prevent overexcitation, a brake that is also part of resilience to stress that protects against depression. Depression then arises when the inhibition is too strong and long lasting - a maladaption, allostatic load, leading to depletion of NA levels in the forebrain. It is suggested that disinhibition by a galanin antagonist may have antidepressant activity by restoring forebrain NA levels. A role of galanin in depression is also supported by a recent candidate gene study, showing that variants in genes for galanin and its three receptors confer increased risk of depression and anxiety in people who experienced childhood adversity or recent negative life events. In summary, galanin, a neuropeptide coexisting in LC neurons, may participate in the mechanism underlying resilience against a serious and common disorder, MDD. Existing and further results may lead to an increased understanding of how this illness develops, which in turn could provide a basis for its treatment.
Collapse
Affiliation(s)
- Tomas Hökfelt
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Swapnali Barde
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Zhi-Qing David Xu
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Neurobiology, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Laboratory of Brain Disorders (Ministry of Science and Technology), Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Eugenia Kuteeva
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Joelle Rüegg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- The Center for Molecular Medicine, Stockholm, Sweden
- Swedish Toxicology Sciences Research Center, Swetox, Södertälje, Sweden
| | - Erwan Le Maitre
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Mårten Risling
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jan Kehr
- Pronexus Analytical AB, Solna, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Robert Ihnatko
- Department of Clinical Chemistry, Linköping University, Linköping, Sweden
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Elvar Theodorsson
- Department of Clinical Chemistry, Linköping University, Linköping, Sweden
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Miklos Palkovits
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - William Deakin
- Neuroscience and Psychiatry Unit, University of Manchester, Manchester, United Kingdom
| | - Gyorgy Bagdy
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
- MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
- NAP 2-SE New Antidepressant Target Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | - Gabriella Juhasz
- Neuroscience and Psychiatry Unit, University of Manchester, Manchester, United Kingdom
- Department of Pharmacodynamics, Semmelweis University, Budapest, Hungary
- SE-NAP2 Genetic Brain Imaging Migraine Research Group, Hungarian Brain Research Program, Semmelweis University, Budapest, Hungary
| | | | - Naguib Mechawar
- Douglas Hospital Research Centre, Verdun, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| | | | - Sven Ove Ögren
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
50
|
Caraci F, Calabrese F, Molteni R, Bartova L, Dold M, Leggio GM, Fabbri C, Mendlewicz J, Racagni G, Kasper S, Riva MA, Drago F. International Union of Basic and Clinical Pharmacology CIV: The Neurobiology of Treatment-resistant Depression: From Antidepressant Classifications to Novel Pharmacological Targets. Pharmacol Rev 2018; 70:475-504. [PMID: 29884653 DOI: 10.1124/pr.117.014977] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Major depressive disorder is one of the most prevalent and life-threatening forms of mental illnesses and a major cause of morbidity worldwide. Currently available antidepressants are effective for most patients, although around 30% are considered treatment resistant (TRD), a condition that is associated with a significant impairment of cognitive function and poor quality of life. In this respect, the identification of the molecular mechanisms contributing to TRD represents an essential step for the design of novel and more efficacious drugs able to modify the clinical course of this disorder and increase remission rates in clinical practice. New insights into the neurobiology of TRD have shed light on the role of a number of different mechanisms, including the glutamatergic system, immune/inflammatory systems, neurotrophin function, and epigenetics. Advances in drug discovery processes in TRD have also influenced the classification of antidepressant drugs and novel classifications are available, such as the neuroscience-based nomenclature that can incorporate such advances in drug development for TRD. This review aims to provide an up-to-date description of key mechanisms in TRD and describe current therapeutic strategies for TRD before examining novel approaches that may ultimately address important neurobiological mechanisms not targeted by currently available antidepressants. All in all, we suggest that drug targeting different neurobiological systems should be able to restore normal function but must also promote resilience to reduce the long-term vulnerability to recurrent depressive episodes.
Collapse
Affiliation(s)
- F Caraci
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - F Calabrese
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - R Molteni
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - L Bartova
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - M Dold
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - G M Leggio
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - C Fabbri
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - J Mendlewicz
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - G Racagni
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - S Kasper
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - M A Riva
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| | - F Drago
- Departments of Drug Sciences (F.Car.) and Biomedical and Biotechnological Sciences, School of Medicine (G.M.L., F.D.), University of Catania, Catania, Italy; Oasi-Research-Institute-IRCCS, Troina, Italy (F.Car.); Departments of Pharmacological and Biomolecular Sciences (F.Cal., G.R., M.A.R.) and Medical Biotechnology and Translational Medicine (R.M.), Università degli Studi di Milano, Milan, Italy; Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria (L.B., M.D., S.K.); Department of Biomedical and NeuroMotor Sciences, University of Bologna, Bologna, Italy (C.F.); and School of Medicine, Universite' Libre de Bruxelles, Bruxelles, Belgium (J.M.)
| |
Collapse
|