1
|
Dubrovskii VN, Maslakova KY, Savchenko EA. Influence of Anticholinesterase Drugs on Activity and Properties of Na +,K +-ATPase in Rat Erythrocytes under Stress Caused by Intense Physical Exercise. Bull Exp Biol Med 2024:10.1007/s10517-024-06202-z. [PMID: 39259465 DOI: 10.1007/s10517-024-06202-z] [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: 11/23/2023] [Indexed: 09/13/2024]
Abstract
We studied the effect of intramuscular injection of physostigmine and neostigmine on Na+,K+-ATPase activity in erythrocytes of rats subjected to intense physical exercise. Both anticholinesterase drugs had a significant effect on the development of the stress response, which was expressed in a decrease in the manifestation of its individual components such as the concentration of ascorbic acid in the adrenal glands, stress-related erythrocyte polycythemia, and LPO indicators. Anticholinesterase drugs reverse the stress-induced decrease in Na+,K+-ATPase activity, as well as changes in its magnesium-dependent properties. There were no changes in the activity of the studied enzyme in the erythrocyte ghosts. We associate the observed differences with the correction of the functions of the cholinergic components of the hypothalamic-pituitary-adrenal axis leading to the development of a hypoergic type stress reaction.
Collapse
|
2
|
Dean B, Scarr E. Common changes in rat cortical gene expression after valproate or lithium treatment particularly affect pre- and post-synaptic pathways that regulate four neurotransmitters systems. World J Biol Psychiatry 2024; 25:54-64. [PMID: 37722808 DOI: 10.1080/15622975.2023.2258972] [Citation(s) in RCA: 2] [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: 06/23/2023] [Accepted: 09/11/2023] [Indexed: 09/20/2023]
Abstract
OBJECTIVES We have postulated that common changes in gene expression after treatment with different therapeutic classes of psychotropic drugs contribute to their common therapeutic mechanisms of action. METHODS To test this hypothesis, we measured levels of cortical coding and non-coding RNA using GeneChip® Rat Exon 1.0 ST Array after treatment with vehicle (chow only), chow containing 1.8 g lithium carbonate/kg (n = 10) or chow containing 12 g sodium valproate/kg (n = 10) for 28 days. Differences in levels of RNA were identified using JMP Genomics 13 and the Panther Gene Ontology Classification System was used to identify potential consequences of RNA. RESULTS Compared to vehicle treatment, levels of cortical RNA for 543 and 583 coding and non-coding RNAs were different after treatment with valproate and lithium, respectively. Moreover, levels of 323 coding and non-coding RNAs were altered in a highly correlated way by treatment with valproate and lithium, changes that would impact on cholinergic, glutamatergic, serotonergic and dopaminergic neurotransmission as well as on voltage gated ion channels. CONCLUSIONS Our study suggests that treating with mood stabilisers cause many common changes in levels of RNA which will impact on CNS function, particularly affecting post-synaptic muscarinic receptor functioning and the release of multiple neurotransmitters.
Collapse
Affiliation(s)
- Brian Dean
- The Molecular Psychiatry Laboratory, The Florey Institute for Neuroscience and Mental Health, Parkville, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Elizabeth Scarr
- The Department of Psychiatry, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
3
|
Wong TS, Li G, Li S, Gao W, Chen G, Gan S, Zhang M, Li H, Wu S, Du Y. G protein-coupled receptors in neurodegenerative diseases and psychiatric disorders. Signal Transduct Target Ther 2023; 8:177. [PMID: 37137892 PMCID: PMC10154768 DOI: 10.1038/s41392-023-01427-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 02/17/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Neuropsychiatric disorders are multifactorial disorders with diverse aetiological factors. Identifying treatment targets is challenging because the diseases are resulting from heterogeneous biological, genetic, and environmental factors. Nevertheless, the increasing understanding of G protein-coupled receptor (GPCR) opens a new possibility in drug discovery. Harnessing our knowledge of molecular mechanisms and structural information of GPCRs will be advantageous for developing effective drugs. This review provides an overview of the role of GPCRs in various neurodegenerative and psychiatric diseases. Besides, we highlight the emerging opportunities of novel GPCR targets and address recent progress in GPCR drug development.
Collapse
Affiliation(s)
- Thian-Sze Wong
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Guangzhi Li
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China
| | - Shiliang Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Wei Gao
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Geng Chen
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Shiyi Gan
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China
| | - Manzhan Zhang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China
| | - Honglin Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 200237, Shanghai, China.
- Innovation Center for AI and Drug Discovery, East China Normal University, 200062, Shanghai, China.
| | - Song Wu
- Institute of Urology, The Affiliated Luohu Hospital of Shenzhen University, Shenzhen University, 518000, Shenzhen, Guangdong, China.
- Department of Urology, South China Hospital, Health Science Center, Shenzhen University, 518116, Shenzhen, Guangdong, China.
| | - Yang Du
- Kobilka Institute of Innovative Drug Discovery, Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 518172, Shenzhen, Guangdong, China.
| |
Collapse
|
4
|
Nabulsi L, Farrell J, McPhilemy G, Kilmartin L, Dauvermann MR, Akudjedu TN, Najt P, Ambati S, Martyn FM, McLoughlin J, Gill M, Meaney J, Morris D, Frodl T, McDonald C, Hallahan B, Cannon DM. Normalization of impaired emotion inhibition in bipolar disorder mediated by cholinergic neurotransmission in the cingulate cortex. Neuropsychopharmacology 2022; 47:1643-1651. [PMID: 35046509 PMCID: PMC9283431 DOI: 10.1038/s41386-022-01268-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/13/2021] [Accepted: 01/04/2022] [Indexed: 11/25/2022]
Abstract
The muscarinic-cholinergic system is involved in the pathophysiology of bipolar disorder (BD), and contributes to attention and the top-down and bottom-up cognitive and affective mechanisms of emotional processing, functionally altered in BD. Emotion processing can be assessed by the ability to inhibit a response when the content of the image is emotional. Impaired regulatory capacity of cholinergic neurotransmission conferred by reduced M2-autoreceptor availability is hypothesized to play a role in elevated salience of negative emotional distractors in euthymic BD relative to individuals with no history of mood instability. Thirty-three euthymic BD type-I (DSM-V-TR) and 50 psychiatrically-healthy controls underwent functional magnetic resonance imaging (fMRI) and an emotion-inhibition paradigm before and after intravenous cholinergic challenge using the acetylcholinesterase inhibitor, physostigmine (1 mg), or placebo. Mood, accuracy, and reaction time on either recognizing or inhibiting a response associated with an image involving emotion and regional functional activation were examined for effects of cholinergic challenge physostigmine relative to placebo, prioritizing any interaction with the diagnostic group. Analyses revealed that (1) at baseline, impaired behavioral performance was associated with lower activation in the anterior cingulate cortex in BD relative to controls during emotion processing; (2) physostigmine (vs. placebo) affected behavioral performance during the inhibition of negative emotions, without altering mood, and increased activation in the posterior cingulate cortex in BD (vs. controls); (3) In BD, lower accuracy observed during emotion inhibition of negative emotions was remediated by physostigmine and was associated with cingulate cortex overactivation. Our findings implicate abnormal regulation of cholinergic neurotransmission in the cingulate cortices in BD, which may mediate exaggerated emotional salience processing, a core feature of BD.
Collapse
Affiliation(s)
- Leila Nabulsi
- Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33, Galway, Ireland. .,Imaging Genetics Center, Mark and Mary Stevens Neuroimaging & Informatics Institute, University of Southern California, Marina del Rey, CA, 90292, USA.
| | - Jennifer Farrell
- grid.6142.10000 0004 0488 0789Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Genevieve McPhilemy
- grid.6142.10000 0004 0488 0789Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Liam Kilmartin
- grid.6142.10000 0004 0488 0789College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| | - Maria R. Dauvermann
- grid.6142.10000 0004 0488 0789Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland ,grid.13097.3c0000 0001 2322 6764Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 8AF UK
| | - Theophilus N. Akudjedu
- grid.6142.10000 0004 0488 0789Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland ,grid.17236.310000 0001 0728 4630Institute of Medical Imaging & Visualisation, Bournemouth University, Bournemouth Gateway Building, St Paul’s Lane, Dorset, BH12 5BB UK
| | - Pablo Najt
- grid.6142.10000 0004 0488 0789Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Srinath Ambati
- grid.6142.10000 0004 0488 0789Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Fiona M. Martyn
- grid.6142.10000 0004 0488 0789Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - James McLoughlin
- grid.6142.10000 0004 0488 0789Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Michael Gill
- grid.8217.c0000 0004 1936 9705Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - James Meaney
- grid.8217.c0000 0004 1936 9705Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Derek Morris
- grid.6142.10000 0004 0488 0789Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Thomas Frodl
- grid.8217.c0000 0004 1936 9705Department of Psychiatry, School of Medicine, Trinity College Dublin, Dublin, Ireland ,Department of Psychiatry and Psychotherapy, Otto-von-Guericke-Universität Magdeburg, University Hospital Magdeburg, Magdeburg, Germany
| | - Colm McDonald
- grid.6142.10000 0004 0488 0789Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Brian Hallahan
- grid.6142.10000 0004 0488 0789Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| | - Dara M. Cannon
- grid.6142.10000 0004 0488 0789Center for Neuroimaging, Cognition and Genomics (NICOG), Clinical Neuroimaging Lab, NCBES Galway Neuroscience Centre, College of Medicine, Nursing, and Health Sciences, National University of Ireland Galway, H91 TK33 Galway, Ireland
| |
Collapse
|
5
|
Moderate Folic Acid Supplementation in Pregnant Mice Results in Altered Sex-Specific Gene Expression in Brain of Young Mice and Embryos. Nutrients 2022; 14:nu14051051. [PMID: 35268026 PMCID: PMC8912750 DOI: 10.3390/nu14051051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/22/2022] [Accepted: 02/26/2022] [Indexed: 02/07/2023] Open
Abstract
Food fortification and increased vitamin intake have led to higher folic acid (FA) consumption by many pregnant women. We showed that FA-supplemented diet in pregnant mice (fivefold higher FA than the recommended level (5xFASD)) led to hyperactivity-like behavior and memory impairment in pups. Disturbed choline/methyl metabolism and altered placental gene expression were identified. The aim of this study was to examine the impact of 5xFASD on the brain at two developmental stages, postnatal day (P) 30 and embryonic day (E) 17.5. Female C57BL/6 mice were fed a control diet or 5xFASD for 1 month before mating. Diets were maintained throughout the pregnancy and lactation until P30 or during pregnancy until E17.5. The 5xFASD led to sex-specific transcription changes in P30 cerebral cortex and E17.5 cerebrum, with microarrays showing a total of 1003 and 623 changes, respectively. Enhanced mRNA degradation was observed in E17.5 cerebrum. Expression changes of genes involved in neurotransmission, neuronal growth and development, and angiogenesis were verified by qRT-PCR; 12 and 15 genes were verified at P30 and E17.5, respectively. Hippocampal collagen staining suggested decreased vessel density in FASD male embryos. This study provides insight into the mechanisms of neurobehavioral alterations and highlights potential deleterious consequences of moderate folate oversupplementation during pregnancy.
Collapse
|
6
|
NRM 2021 Abstract Booklet. J Cereb Blood Flow Metab 2021; 41:11-309. [PMID: 34905986 PMCID: PMC8851538 DOI: 10.1177/0271678x211061050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
7
|
Tolaymat M, Sundel MH, Alizadeh M, Xie G, Raufman JP. Potential Role for Combined Subtype-Selective Targeting of M 1 and M 3 Muscarinic Receptors in Gastrointestinal and Liver Diseases. Front Pharmacol 2021; 12:786105. [PMID: 34803723 PMCID: PMC8600121 DOI: 10.3389/fphar.2021.786105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/19/2021] [Indexed: 01/17/2023] Open
Abstract
Despite structural similarity, the five subtypes comprising the cholinergic muscarinic family of G protein-coupled receptors regulate remarkably diverse biological functions. This mini review focuses on the closely related and commonly co-expressed M1R and M3R muscarinic acetylcholine receptor subtypes encoded respectively by CHRM1 and CHRM3. Activated M1R and M3R signal via Gq and downstream initiate phospholipid turnover, changes in cell calcium levels, and activation of protein kinases that alter gene transcription and ultimately cell function. The unexpectedly divergent effects of M1R and M3R activation, despite similar receptor structure, distribution, and signaling, are puzzling. To explore this conundrum, we focus on the gastrointestinal (GI) tract and liver because abundant data identify opposing effects of M1R and M3R activation on the progression of gastric, pancreatic, and colon cancer, and liver injury and fibrosis. Whereas M3R activation promotes GI neoplasia, M1R activation appears protective. In contrast, in murine liver injury models, M3R activation promotes and M1R activation mitigates liver fibrosis. We analyze these findings critically, consider their therapeutic implications, and review the pharmacology and availability for research and therapeutics of M1R and M3R-selective agonists and antagonists. We conclude by considering gaps in knowledge and other factors that hinder the application of these drugs and the development of new agents to treat GI and liver diseases.
Collapse
Affiliation(s)
- Mazen Tolaymat
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Margaret H Sundel
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Madeline Alizadeh
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Guofeng Xie
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States.,VA Maryland Healthcare System, Baltimore, MD, United States.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jean-Pierre Raufman
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Maryland School of Medicine, Baltimore, MD, United States.,VA Maryland Healthcare System, Baltimore, MD, United States.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, United States.,Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
8
|
Transcriptome-wide association study of treatment-resistant depression and depression subtypes for drug repurposing. Neuropsychopharmacology 2021; 46:1821-1829. [PMID: 34158615 PMCID: PMC8357803 DOI: 10.1038/s41386-021-01059-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/19/2021] [Accepted: 06/03/2021] [Indexed: 12/17/2022]
Abstract
Major depressive disorder (MDD) is the single largest contributor to global disability and up to 20-30% of patients do not respond to at least two antidepressants (treatment-resistant depression, TRD). This study leveraged imputed gene expression in TRD to perform a drug repurposing analysis. Among those with MDD, we defined TRD as having at least two antidepressant switches according to primary care records in UK Biobank (UKB). We performed a transcriptome-wide association study (TWAS) of TRD (n = 2165) vs healthy controls (n = 11,188) using FUSION and gene expression levels from 21 tissues. We identified compounds with opposite gene expression signatures (ConnectivityMap data) compared to our TWAS results using the Kolmogorov-Smirnov test, Spearman and Pearson correlation. As symptom patterns are routinely assessed in clinical practice and could be used to provide targeted treatments, we identified MDD subtypes associated with TRD in UKB and analysed them using the same pipeline described for TRD. Anxious MDD (n = 14,954) and MDD with weight gain (n = 4697) were associated with TRD. In the TWAS, two genes were significantly dysregulated (TMEM106B and ATP2A1 for anxious and weight gain MDD, respectively). A muscarinic receptor antagonist was identified as top candidate for repurposing in TRD; inhibition of heat shock protein 90 was the main mechanism of action identified for anxious MDD, while modulators of metabolism such as troglitazone showed promising results for MDD with weight gain. This was the first TWAS of TRD and associated MDD subtypes. Our results shed light on possible pharmacological approaches in individuals with difficult-to-treat depression.
Collapse
|
9
|
Halpert G, Watad A, Tsur AM, Dotan A, Quiros-Lim HE, Heidecke H, Gilburd B, Haik J, Levy Y, Blank M, Amital H, Shoenfeld Y. Autoimmune dysautonomia in women with silicone breast implants. J Autoimmun 2021; 120:102631. [PMID: 33799099 DOI: 10.1016/j.jaut.2021.102631] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/07/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022]
Abstract
IMPORTANCE AND OBJECTIVES There is unmet medical need to understand the pathogenic mechanism of the panoply of clinical manifestations associated with silicone breast implants (SBIs) such as severe fatigue, widespread pain, palpitations, dry mouth and eyes, depression, hearing loss etc. We aimed to determine whether autoantibodies against the autonomic nervous system receptors can explain the enigmatic and subjective clinical manifestation reported by women with SBIs. RESULTS Circulating level of autoantibodies against G protein-coupled receptors (GPCRs) of the autonomic nervous system (adrenergic, muscarinic, endothelin and angiotensin receptors) have been evaluated in symptomatic women with SBIs using an ELISA method. These women with SBIs addressed our clinic due to various subjective and autonomic-related manifestations such as chronic severe fatigue, cognitive impairment, widespread pain, memory loss, sleep disorders, palpitations, depression, hearing abnormalities etc. We report for the first time, a significant reduction in the sera level of anti-β1 adrenergic receptor (p < 0.001), anti-angiotensin II type 1 receptor (p < 0.001) and anti-endothelin receptor type A (p = 0.001) autoantibodies in women with SBIs (n = 93) as compared with aged matched healthy women (n = 36). Importantly, anti-β1 adrenergic receptor autoantibody was found to significantly correlate with autonomic-related manifestations such as: sleep disorders and depression in women with SBIs. CONCLUSIONS Chronic immune stimulation by silicone material may lead to an autoimmune dysautonomia in a subgroup of potentially genetically susceptible women with SBIs. The appearance of autoantibodies against GPCRs of the autonomic nervous system serve as an explanation for the subjective autonomic-related manifestations reported in women with SBIs.
Collapse
Affiliation(s)
- Gilad Halpert
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat- Gan, 52621, Israel; Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Abdulla Watad
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat- Gan, 52621, Israel; Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Medicine 'B' and Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Israel
| | - Avishai M Tsur
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat- Gan, 52621, Israel; Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Israel Defense Forces, Medical Corps, Tel Hashomer, Ramat Gan, Israel; Department of Military Medicine, Hebrew University of Jerusalem Faculty of Medicine, Jerusalem, Israel
| | - Arad Dotan
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat- Gan, 52621, Israel
| | - Hector Enrique Quiros-Lim
- Department of Plastic and Reconstructive Surgery. The Chaim Sheba Medical Center at Tel Hashomer. Ramat Gan. Israel
| | | | - Boris Gilburd
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat- Gan, 52621, Israel; Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Josef Haik
- Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Plastic and Reconstructive Surgery. The Chaim Sheba Medical Center at Tel Hashomer. Ramat Gan. Israel; College of Health and Medicine. University of Tasmania, Sydney, NSW, Australia; Institute for Health Research. University of Notre Dame, Fremantle, Australia
| | - Yair Levy
- Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Medicine E, Meir Medical Center, Kfar Saba, Israel
| | - Miri Blank
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat- Gan, 52621, Israel; Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Howard Amital
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat- Gan, 52621, Israel; Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Medicine 'B' and Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Ramat- Gan, 52621, Israel; Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
10
|
Sanches M, Quevedo J, Soares JC. New agents and perspectives in the pharmacological treatment of major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110157. [PMID: 33159975 PMCID: PMC7750246 DOI: 10.1016/j.pnpbp.2020.110157] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/19/2020] [Accepted: 10/27/2020] [Indexed: 12/28/2022]
Abstract
Despite the important advances in the understanding of the pathophysiology of MDD, a large proportion of depressed patients do not respond well to currently available pharmacological agents. The present review focuses on new targets and future directions in the pharmacological treatment of MDD. Novel agents and their efficacy in the treatment of depression are discussed, with a focus on the respectively target pathophysiological pathways and the level of available evidence. Although it is expected that classic antidepressants will remain the cornerstone of MDD treatment, at least for the near future, a large number of novel compounds is currently under investigation as for their efficacy in the treatment of MDD, many of which with promising results.
Collapse
Affiliation(s)
- Marsal Sanches
- UT Health Center of Excellence on Mood Disorders, Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| | - Joao Quevedo
- UT Health Center of Excellence on Mood Disorders, Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Translational Psychiatry Program, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA; Translational Psychiatry Laboratory, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Jair C Soares
- UT Health Center of Excellence on Mood Disorders, Faillace Department of Psychiatry & Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| |
Collapse
|
11
|
Functional coupling of M 1 muscarinic acetylcholine receptor to Gα q/11 in dorsolateral prefrontal cortex from patients with psychiatric disorders: a postmortem study. Eur Arch Psychiatry Clin Neurosci 2020; 270:869-880. [PMID: 31807862 DOI: 10.1007/s00406-019-01088-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 11/23/2019] [Indexed: 01/14/2023]
Abstract
Accumulating studies have implicated intracellular signaling through muscarinic acetylcholine receptors (mAChRs) in psychiatric illness. In the present study, carbamylcholine chloride (carbachol)-induced Gαi/o and Gαq/11 activation was identified in postmortem human prefrontal cortical membranes. The following two sample cohorts were used: subjects [1], consisting of 40 controls without neuropsychiatric disorders, and subjects [2], consisting of 20 with bipolar disorder (BP), 20 major depressive disorder (MDD), 20 schizophrenia, and 20 controls, strictly sex- and age-matched. Carbachol-stimulated [35S]GTPγS binding to human brain membranes was assessed by the two methods, i.e., conventional method using filtration techniques (Gαi/o activation coupled to M2/M4 mAChRs) applied to subjects [1], and [35S]GTPγS binding/immuno precipitation assay (Gαq/11 activation coupled to M1 mAChR) applied to subjects [1] and [2]. The concentration eliciting the half-maximal effect (EC50), maximum percent increase (%Emax), and slope factor were obtained from concentration-response curve of carbachol-induced Gαi/o and Gαq/11 activation. The pEC50 values of both carbachol-induced Gαi/o and Gαq/11 activations in subjects [1] were significantly correlated, though its implications or underlying molecular processes are unclear. The results of M1 mAChR-mediated Gαq/11 activation in subjects [2] indicated no significant disorder-specific alterations. However, the distribution patterns of the pEC50 values showed unequal variances among the groups. There was a significant inverse correlation between the %Emax values and the pEC50 values in subjects with schizophrenia, but not in those with BP or MDD, or controls. These data support the notion that schizophrenia patients consist of biologically heterogeneous subgroups with respect to M1 mAChR-mediated signaling pathways.
Collapse
|
12
|
Na +, K +-ATPase α Isoforms and Endogenous Cardiac Steroids in Prefrontal Cortex of Bipolar Patients and Controls. Int J Mol Sci 2020; 21:ijms21165912. [PMID: 32824628 PMCID: PMC7460572 DOI: 10.3390/ijms21165912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/05/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023] Open
Abstract
Bipolar disorder is a chronic multifactorial psychiatric illness that affects the mood, cognition, and functioning of about 1–2% of the world’s population. Its biological basis is unknown, and its treatment is unsatisfactory. The α1, α2, and α3 isoforms of the Na+, K+-ATPase, an essential membrane transporter, are vital for neuronal and glial function. The enzyme and its regulators, endogenous cardiac steroids like ouabain and marinobufagenin, are implicated in neuropsychiatric disorders, bipolar disorder in particular. Here, we address the hypothesis that the α isoforms of the Na+, K+-ATPase and its regulators are altered in the prefrontal cortex of bipolar disease patients. The α isoforms were determined by Western blot and ouabain and marinobufagenin by specific and sensitive immunoassays. We found that the α2 and α3 isoforms were significantly higher and marinobufagenin levels were significantly lower in the prefrontal cortex of the bipolar disease patients compared with those in the control. A positive correlation was found between the levels of the three α isoforms in all samples and between the α1 isoform and ouabain levels in the controls. These results are in accordance with the notion that the Na+, K+-ATPase-endogenous cardiac steroids system is involved in bipolar disease and suggest that it may be used as a target for drug development.
Collapse
|
13
|
Puryear CB, Brooks J, Tan L, Smith K, Li Y, Cunningham J, Todtenkopf MS, Dean RL, Sanchez C. Opioid receptor modulation of neural circuits in depression: What can be learned from preclinical data? Neurosci Biobehav Rev 2020; 108:658-678. [DOI: 10.1016/j.neubiorev.2019.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/02/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
|
14
|
Boiko AS, Ivanova SA, Pozhidaev IV, Freidin MB, Osmanova DZ, Fedorenko OY, Semke AV, Bokhan NA, Wilffert B, Loonen AJM. Pharmacogenetics of tardive dyskinesia in schizophrenia: The role of CHRM1 and CHRM2 muscarinic receptors. World J Biol Psychiatry 2020; 21:72-77. [PMID: 30623717 DOI: 10.1080/15622975.2018.1548780] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Objectives: Acetylcholine M (muscarinic) receptors are possibly involved in tardive dyskinesia (TD). The authors tried to verify this hypothesis by testing for possible associations between two muscarinic receptor genes (CHRM1 and CHRM2) polymorphisms and TD in patients with schizophrenia.Methods: A total of 472 patients with schizophrenia were recruited. TD was assessed cross-sectionally using the Abnormal Involuntary Movement Scale. Fourteen allelic variants of CHRM1 and CHRM2 were genotyped using Applied Biosystems amplifiers (USA) and the MassARRAY System by Agena Bioscience.Results: The prevalence of the rs1824024*GG genotype of the CHRM2 gene was lower in TD patients compared to the group without it (χ2 = 6.035, p = 0.049). This suggested that this genotype has a protective effect for the development of TD (OR = 0.4, 95% CI: 0.19-0.88). When age, gender, duration of schizophrenia and dosage of antipsychotic treatment were added as covariates in regression analysis, the results did not reach statistical significance.Conclusions: This study did identify associations between CHRM2 variations and TD; the results of logistic regression analysis with covariates suggest that the association is, however, likely to be secondary to other concomitant factors.
Collapse
Affiliation(s)
- Anastasiia S Boiko
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation
| | - Svetlana A Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation.,National Research Tomsk Polytechnic University, Tomsk, Russian Federation
| | - Ivan V Pozhidaev
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation.,National Research Tomsk State University, Tomsk, Russian Federation
| | - Maxim B Freidin
- Department of Twin Research and Genetic Epidemiology, School of Live Course Sciences, King's College London, London, United Kingdom.,Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation
| | - Diana Z Osmanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation.,National Research Tomsk State University, Tomsk, Russian Federation
| | - Olga Yu Fedorenko
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation.,National Research Tomsk Polytechnic University, Tomsk, Russian Federation
| | - Arkadyi V Semke
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation
| | - Nikolay A Bokhan
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation.,National Research Tomsk State University, Tomsk, Russian Federation
| | - Bob Wilffert
- Groningen Research Institute of Pharmacy, Unit of PharmacoTherapy, -Epidemiology & -Economics, University of Groningen, Groningen, the Netherlands.,Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Anton J M Loonen
- Groningen Research Institute of Pharmacy, Unit of PharmacoTherapy, -Epidemiology & -Economics, University of Groningen, Groningen, the Netherlands.,GGZ WNB, Mental health hospital, Bergen op Zoom, The Netherlands
| |
Collapse
|
15
|
Prus AJ, Porter JH. The Discriminative Stimulus Properties of Drugs Used to Treat Depression and Anxiety. Curr Top Behav Neurosci 2019; 39:213-241. [PMID: 27352389 DOI: 10.1007/7854_2016_27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Drug discrimination is a powerful tool for evaluating the stimulus effects of psychoactive drugs and for linking these effects to pharmacological mechanisms. This chapter reviews the primary findings from drug discrimination studies of antidepressant and anxiolytic drugs, including novel pharmacological mechanisms. The stimulus properties revealed from these animal studies largely correspond to the receptor affinities of antidepressant and anxiolytic drugs, indicating that subjective effects may correspond to either therapeutic or side effects of these medications. We discuss drug discrimination findings concerning adjunctive medications and novel pharmacologic strategies in antidepressant and anxiolytic research. Future directions for drug discrimination work include an urgent need to explore the subjective effects of medications in animal models, to better understand shifts in stimulus sensitivity during prolonged treatments, and to further characterize stimulus effects in female subjects. We conclude that drug discrimination is an informative preclinical procedure that reveals the interoceptive effects of pharmacological mechanisms as they relate to behaviors that are not captured in other preclinical models.
Collapse
Affiliation(s)
- Adam J Prus
- Department of Psychology, Northern Michigan University, Marquette, MI, USA.
| | - Joseph H Porter
- Department of Psychology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
16
|
Witkin JM, Martin AE, Golani LK, Xu NZ, Smith JL. Rapid-acting antidepressants. ADVANCES IN PHARMACOLOGY 2019; 86:47-96. [DOI: 10.1016/bs.apha.2019.03.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
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
|
18
|
Lichtstein D, Ilani A, Rosen H, Horesh N, Singh SV, Buzaglo N, Hodes A. Na⁺, K⁺-ATPase Signaling and Bipolar Disorder. Int J Mol Sci 2018; 19:E2314. [PMID: 30087257 PMCID: PMC6121236 DOI: 10.3390/ijms19082314] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 02/07/2023] Open
Abstract
Bipolar disorder (BD) is a severe and common chronic mental illness characterized by recurrent mood swings between depression and mania. The biological basis of the disease is poorly understood and its treatment is unsatisfactory. Although in past decades the "monoamine hypothesis" has dominated our understanding of both the pathophysiology of depressive disorders and the action of pharmacological treatments, recent studies focus on the involvement of additional neurotransmitters/neuromodulators systems and cellular processes in BD. Here, evidence for the participation of Na⁺, K⁺-ATPase and its endogenous regulators, the endogenous cardiac steroids (ECS), in the etiology of BD is reviewed. Proof for the involvement of brain Na⁺, K⁺-ATPase and ECS in behavior is summarized and it is hypothesized that ECS-Na⁺, K⁺-ATPase-induced activation of intracellular signaling participates in the mechanisms underlying BD. We propose that the activation of ERK, AKT, and NFκB, resulting from ECS-Na⁺, K⁺-ATPase interaction, modifies neuronal activity and neurotransmission which, in turn, participate in the regulation of behavior and BD. These observations suggest Na⁺, K⁺-ATPase-mediated signaling is a potential target for drug development for the treatment of BD.
Collapse
Affiliation(s)
- David Lichtstein
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| | - Asher Ilani
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| | - Haim Rosen
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| | - Noa Horesh
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| | - Shiv Vardan Singh
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| | - Nahum Buzaglo
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| | - Anastasia Hodes
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
19
|
Fornaro M, Kardash L, Novello S, Fusco A, Anastasia A, De Berardis D, Perna G, Carta MG. Progress in bipolar disorder drug design toward the development of novel therapeutic targets: a clinician's perspective. Expert Opin Drug Discov 2018; 13:221-228. [PMID: 29357703 DOI: 10.1080/17460441.2018.1428554] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Bipolar disorder (BD) is a considerable burden to the affected individual. The need for novel drug targets and improved drug design (DD) in BD is therefore clear. Areas covered: The following article provides a brief, narrative, clinician-oriented overview of the most promising novel pharmacological targets for BD along with a concise overview regarding the general DD process and the unmet needs relevant to BD. Expert opinion: A number of novel potential drug targets have been investigated. With the notable exception of the kynurenine pathway, available evidence is too scarce to highlight a definitive roadmap for forthcoming DD in BD. BD itself may present with different facets, as it is a polymorphic clinical spectrum. Therefore, promoting clinical-case stratification should be based on precision medicine, rather than on novel biological targets. Furthermore, the full release of raw study data to the scientific community and the development of uniform clinical trial standards (including more realistic outcomes) should be promoted to facilitate the DD process in BD.
Collapse
Affiliation(s)
- Michele Fornaro
- a Department of Neuroscience, Reproductive Sciences and Odontostomatology, Section of Psychiatry, University School of Medicine , "Federico II" University of Naples , Italy
| | - Lubna Kardash
- b Department of Internal Medicine , Wayne State University , Detroit , MI , USA
| | - Stefano Novello
- a Department of Neuroscience, Reproductive Sciences and Odontostomatology, Section of Psychiatry, University School of Medicine , "Federico II" University of Naples , Italy
| | - Andrea Fusco
- a Department of Neuroscience, Reproductive Sciences and Odontostomatology, Section of Psychiatry, University School of Medicine , "Federico II" University of Naples , Italy
| | - Annalisa Anastasia
- a Department of Neuroscience, Reproductive Sciences and Odontostomatology, Section of Psychiatry, University School of Medicine , "Federico II" University of Naples , Italy
| | - Domenico De Berardis
- c Department of Mental Health, Psychiatric Service of Diagnosis and Treatment , National Health Service, Hospital "G. Mazzini" , Teramo , Italy
| | - Giampaolo Perna
- d Department of Psychiatry and Neuropsychology , Maastricht University , Maastricht , Netherlands.,e Department of Clinical Neurosciences , FoRiPsi, Hermanas Hospitalarias-Villa San Benedetto Menni Hospital , Como , Italy.,f Department of Psychiatry and Behavioral Sciences, Leonard Miller School of Medicine , University of Miami , Miami , FL , USA
| | | |
Collapse
|
20
|
Yu H, Li M, Zhou D, Lv D, Liao Q, Lou Z, Shen M, Wang Z, Li M, Xiao X, Zhang Y, Wang C. Vesicular glutamate transporter 1 (VGLUT1)-mediated glutamate release and membrane GluA1 activation is involved in the rapid antidepressant-like effects of scopolamine in mice. Neuropharmacology 2017; 131:209-222. [PMID: 29274366 DOI: 10.1016/j.neuropharm.2017.12.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 12/12/2017] [Accepted: 12/17/2017] [Indexed: 12/19/2022]
Abstract
Emerging data have identified certain drugs such as scopolamine as rapidly acting antidepressants for major depressive disorder (MDD) that increase glutamate release and induce neurotrophic factors through α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) activation in rodent models. However, little research has addressed the direct mechanisms of scopolamine on AMPAR activation or vesicular glutamate transporter 1 (VGLUT1)-mediated glutamate release in the prefrontal cortex (PFC) of mice. Herein, using a chronic unpredictable stress (CUS) paradigm, acute treatment with scopolamine rapidly reversed stress-induced depression-like behaviors in mice. Our results showed that CUS-induced depression-like behaviors, accompanied by a decrease in membrane AMPAR subunit 1 (GluA1), phosphorylated GluA1 Ser845 (pGluA1 Ser845), brain-derived neurotrophic factor (BDNF) and VGF (non-acronymic) and an increase in bicaudal C homolog 1 gene (BICC1) in the PFC of mice, and these biochemical and behavioral abnormalities were ameliorated by acute scopolamine treatments. However, pharmacological block of AMPAR by NBQX infusion into the PFC significantly abolished these effects of scopolamine. In addition, knock down of VGLUT1 by lentiviral-mediated RNA interference in the PFC of mice was sufficient to induce depression-like phenotype, to decrease extracellular glutamate accumulation and to cause similar molecular changes with CUS in mice. Remarkably, VGLUT1 knockdown alleviated the rapid antidepressant-like actions of scopolamine and the effects of scopolamine on membrane GluA1-mediated BDNF, VGF and BICC1 changes. Altogether, our findings suggest that VGLUT1-mediated glutamate release and membrane GluA1 activation may play a critical role in the rapid-acting antidepressant-like effects of scopolamine in mice.
Collapse
Affiliation(s)
- Hanjie Yu
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China
| | - Mengmeng Li
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China
| | - Dongsheng Zhou
- Ningbo Kangning Hospital, Ningbo, Zhejiang 315201, China
| | - Dan Lv
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China
| | - Qi Liao
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China
| | - Zhongze Lou
- Department of Psychosomatic Medicine, Ningbo First Hospital, 59 Liuting Str., Ningbo, Zhejiang 315010, China
| | - Mengxin Shen
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China
| | - Zhen Wang
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, 32 East Jiao-Chang Rd, Kunming, Yunnan 650223, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, 32 East Jiao-Chang Rd, Kunming, Yunnan 650223, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanhua Zhang
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China
| | - Chuang Wang
- Ningbo Key Laboratory of Behavioral Neuroscience, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China; Department of Physiology and Pharmacology, Ningbo University School of Medicine, 818 Fenghua Road, Ningbo, Zhejiang 315211, China.
| |
Collapse
|
21
|
Hernández-Abrego A, Vázquez-Gómez E, García-Colunga J. Effects of the antidepressant mirtazapine and zinc on nicotinic acetylcholine receptors. Neurosci Lett 2017; 665:246-251. [PMID: 29225093 DOI: 10.1016/j.neulet.2017.12.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/29/2017] [Accepted: 12/06/2017] [Indexed: 12/23/2022]
Abstract
Nicotinic acetylcholine receptors (nAChRs) and zinc are associated with regulation of mood and related disorders. In addition, several antidepressants inhibit muscle and neuronal nAChRs and zinc potentiates inhibitory actions of them. Moreover, mirtazapine (a noradrenergic, serotonergic and histaminergic antidepressant) inhibits muscarinic AChRs and its effects on nAChRs are unknown. Therefore, we studied the modulation of muscle α1β1γd nAChRs expressed in oocytes and native α7-containing nAChRs in hippocampal interneurons by mirtazapine and/or zinc, using voltage-clamp techniques. The currents elicited by ACh in oocytes (at -60 mV) were similarly inhibited by mirtazapine in the absence and presence of 100 μM zinc (IC50 ∼15 μM); however, the ACh-induced currents were stronger inhibited with 20 and 50 μM mirtazapine in the presence of zinc. Furthermore, the potentiation of ACh-induced current by zinc in the presence of 5 μM mirtazapine was 1.48 ± 0.06, and with 50 μM mirtazapine zinc potentiation did not occur. Interestingly, in stratum radiatum interneurons (at -70 mV), 20 μM mirtazapine showed less inhibition of the current elicited by choline (Ch) than at 10 μM (0.81 ± 0.02 and 0.74 ± 0.02 of the Ch-induced current, respectively). Finally, the inhibitory effects of mirtazapine depended on membrane potential: 0.81 ± 0.02 and 0.56 ± 0.05 of the control Ch-induced current at -70 and -20 mV, respectively. These results indicate that mirtazapine interacts with muscle and neuronal nAChRs, possibly into the ion channel; that zinc may increase the sensitivity of nAChRs to mirtazapine; and that mirtazapine decreases the sensitivity of nAChRs to zinc.
Collapse
Affiliation(s)
- Andy Hernández-Abrego
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla, Querétaro 76230, México
| | - Elizabeth Vázquez-Gómez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla, Querétaro 76230, México
| | - Jesús García-Colunga
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla 3001, Juriquilla, Querétaro 76230, México.
| |
Collapse
|
22
|
Buoli M, Esposito CM, Godio M, Caldiroli A, Serati M, Altamura AC. Have antipsychotics a different speed of action in the acute treatment of mania? A single-blind comparative study. J Psychopharmacol 2017; 31:1537-1543. [PMID: 28462607 DOI: 10.1177/0269881117705098] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Available antipsychotics show different efficacy on manic symptoms of bipolar patients, but few studies have investigated the speed of action of the various compounds. For this reason, purpose of the present paper was to compare antipsychotic mono-therapies in terms of speed of action in a sample of manic bipolar patients. In total, 155 bipolar patients, treated with antipsychotic mono-therapy and followed-up in Inpatient Psychiatry Clinic of University of Milan, were included in this single-blind comparative study. Clinical response was defined as a reduction of Young Mania Rating Scale (YMRS) scores ⩾50%, while remission as a YMRS score <10. After 4 days patients who had been treated with asenapine, were more likely to have achieved a clinical response than those in treatment with haloperidol ( p = 0.001). After 7 days, a more frequent clinical response was achieved by patients treated with asenapine than those who had been treated with haloperidol ( p < 0.001) or olanzapine ( p = 0.047). Asenapine appears to be faster in determining treatment response in manic patients compared with haloperidol and less markedly with olanzapine.
Collapse
Affiliation(s)
- Massimiliano Buoli
- 1 Department of Psychiatry, University of Milan, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Cecilia Maria Esposito
- 1 Department of Psychiatry, University of Milan, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Godio
- 2 Cantonal Psychiatric Clinic, Mendrisio, Switzerland
| | - Alice Caldiroli
- 1 Department of Psychiatry, University of Milan, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marta Serati
- 1 Department of Psychiatry, University of Milan, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - A Carlo Altamura
- 1 Department of Psychiatry, University of Milan, Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
23
|
Smail MA, Soles JL, Karwoski TE, Rubin RT, Rhodes ME. Sexually diergic hypothalamic-pituitary-adrenal axis responses to selective and non-selective muscarinic antagonists prior to cholinergic stimulation by physostigmine in rats. Brain Res Bull 2017; 137:23-34. [PMID: 29122691 DOI: 10.1016/j.brainresbull.2017.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 10/21/2017] [Accepted: 11/02/2017] [Indexed: 01/02/2023]
Abstract
Central cholinergic systems regulate the hypothalamic-pituitary-adrenal (HPA) axis differentially in males and females (sexual diergism). We previously investigated the role of muscarinic receptors in this regulation by administering physostigmine (PHYSO), an acetylcholinesterase inhibitor, to male and female rats pretreated with scopolamine (SCOP), a nonselective muscarinic antagonist. SCOP pretreatment enhanced adrenocorticotropic hormone (ACTH) and corticosterone (CORT) responses in both sexes, but males had greater ACTH responses while females had greater CORT responses. In the present study, we further explored the role of muscarinic receptor subtypes in HPA axis regulation by administering PHYSO to male and female rats following SCOP or various doses of either the M1 or the M2 selective muscarinic receptor antagonists, pirenzepine (PIREN) or methoctramine (METHO). Blood was sampled before and at multiple times after PHYSO. ACTH and CORT were determined by highly specific immunoassays. M1 antagonism by PIREN prior to PHYSO resulted in sustained, dose-dependent increases in ACTH and CORT: ACTH responses were similar in both sexes, and CORT responses were greater in females. M2 antagonism by METHO prior to PHYSO resulted in overall decreases in ACTH and CORT: ACTH and CORT responses were higher in females but lower in both sexes than the hormone responses following PIREN or SCOP pretreatment. Area under the curve analyses supported these findings. These results suggest that specific muscarinic receptor subtypes differentially influence the HPA axis in a sexually diergic manner.
Collapse
Affiliation(s)
- Marissa A Smail
- Department of Biology, Saint Vincent College, Latrobe, PA, United States
| | - Jessica L Soles
- Department of Biology, Saint Vincent College, Latrobe, PA, United States
| | - Tracy E Karwoski
- Center for Neurosciences Research, Allegheny General Hospital, Pittsburgh, PA, United States
| | - Robert T Rubin
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Michael E Rhodes
- Department of Biology, Saint Vincent College, Latrobe, PA, United States.
| |
Collapse
|
24
|
Sakata K, Overacre AE. Promoter IV-BDNF deficiency disturbs cholinergic gene expression of CHRNA5, CHRM2, and CHRM5: effects of drug and environmental treatments. J Neurochem 2017; 143:49-64. [PMID: 28722769 DOI: 10.1111/jnc.14129] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/05/2017] [Accepted: 07/13/2017] [Indexed: 11/29/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) promotes maturation of cholinergic neurons. However, how activity-dependent BDNF expression affects specific cholinergic gene expression remains unclear. This study addressed this question by determining mRNA levels of 22 acetylcholine receptor subunits, the choline transporter (CHT), and the choline acetyltransferase (ChAT) in mice deficient in activity-dependent BDNF via promoter IV (KIV) and control wild-type mice. Quantitative RT-PCR revealed significant reductions in nicotinic acetylcholine receptor alpha 5 (CHRNA5) in the frontal cortex and hippocampus and M5 muscarinic acetylcholine receptor (CHRM5) in the hippocampus, but significant increases in M2 muscarinic acetylcholine receptor (CHRM2) in the frontal cortex of KIV mice compared to wild-type mice. Three-week treatments with fluoxetine, phenelzine, duloxetine, imipramine, or an enriched environment treatment (EET) did not affect the altered expression of these genes except that EET increased CHRNA5 levels only in KIV frontal cortex. EET also increased levels of CHRNA7, CHT, and ChAT, again only in the KIV frontal cortex. The imipramine treatment was most prominent among the four antidepressants; it up-regulated hippocampal CHRM2 and frontal cortex CHRM5 in both genotypes, and frontal cortex CHRNA7 only in KIV mice. To the best of our knowledge, this is the first evidence that BDNF deficiency disturbs expression of CHRNA5, CHRM2, and CHRM5. Our results suggest that promoter IV-BDNF deficiency - which occurs under chronic stress - causes cholinergic dysfunctions via these receptors. EET is effective on CHRNA5, while its compensatory induction of other cholinergic genes or drugs targeting CHRNA5, CHRM2, and CHRM5 may become an alternative strategy to reverse these BDNF-linked cholinergic dysfunctions.
Collapse
Affiliation(s)
- Kazuko Sakata
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Abigail E Overacre
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
25
|
Blanco-Silvente L, Castells X, Saez M, Barceló MA, Garre-Olmo J, Vilalta-Franch J, Capellà D. Discontinuation, Efficacy, and Safety of Cholinesterase Inhibitors for Alzheimer's Disease: a Meta-Analysis and Meta-Regression of 43 Randomized Clinical Trials Enrolling 16 106 Patients. Int J Neuropsychopharmacol 2017; 20:519-528. [PMID: 28201726 PMCID: PMC5492783 DOI: 10.1093/ijnp/pyx012] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/17/2017] [Accepted: 02/09/2017] [Indexed: 01/10/2023] Open
Abstract
Background We investigated the effect of cholinesterase inhibitors on all-cause discontinuation, efficacy and safety, and the effects of study design-, intervention-, and patient-related covariates on the risk-benefit of cholinesterase inhibitors for Alzheimer's disease. Methods A systematic review and meta-analysis of randomized placebo-controlled clinical trials comparing cholinesterase inhibitors and placebo was performed. The effect of covariates on study outcomes was analysed by means of meta-regression using a Bayesian framework. Results Forty-three randomized placebo-controlled clinical trials involving 16106 patients were included. All-cause discontinuation was higher with cholinesterase inhibitors (OR = 1.66), as was discontinuation due to adverse events (OR=1.75). Cholinesterase inhibitors improved cognitive function (standardized mean difference = 0.38), global symptomatology (standardized mean difference=0.28) and functional capacity (standardized mean difference=0.16) but not neuropsychiatric symptoms. Rivastigmine was associated with a poorer outcome on all-cause discontinuation (Diff OR = 1.66) and donepezil with a higher efficacy on global change (Diff standardized mean difference = 0.41). The proportion of patients with serious adverse events decreased with age (Diff OR = -0.09). Mortality was lower with cholinesterase inhibitors than with placebo (OR = 0.65). Conclusion While cholinesterase inhibitors show a poor risk-benefit relationship as indicated by mild symptom improvement and a higher than placebo all-cause discontinuation, a reduction of mortality was suggested. Intervention- and patient-related factors modify the effect of cholinesterase inhibitors in patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Lídia Blanco-Silvente
- TransLab Research Group, and Department of Medical Sciences, University of Girona, Spain; Research Group on Statistics, Econometrics and Health, University of Girona, Spain; CIBER of Epidemiology and Public Health, Spain; Girona Biomedical Research Institute, Salt, Spain
| | - Xavier Castells
- TransLab Research Group, and Department of Medical Sciences, University of Girona, Spain; Research Group on Statistics, Econometrics and Health, University of Girona, Spain; CIBER of Epidemiology and Public Health, Spain; Girona Biomedical Research Institute, Salt, Spain
| | - Marc Saez
- TransLab Research Group, and Department of Medical Sciences, University of Girona, Spain; Research Group on Statistics, Econometrics and Health, University of Girona, Spain; CIBER of Epidemiology and Public Health, Spain; Girona Biomedical Research Institute, Salt, Spain
| | - Maria Antònia Barceló
- TransLab Research Group, and Department of Medical Sciences, University of Girona, Spain; Research Group on Statistics, Econometrics and Health, University of Girona, Spain; CIBER of Epidemiology and Public Health, Spain; Girona Biomedical Research Institute, Salt, Spain
| | - Josep Garre-Olmo
- TransLab Research Group, and Department of Medical Sciences, University of Girona, Spain; Research Group on Statistics, Econometrics and Health, University of Girona, Spain; CIBER of Epidemiology and Public Health, Spain; Girona Biomedical Research Institute, Salt, Spain
| | - Joan Vilalta-Franch
- TransLab Research Group, and Department of Medical Sciences, University of Girona, Spain; Research Group on Statistics, Econometrics and Health, University of Girona, Spain; CIBER of Epidemiology and Public Health, Spain; Girona Biomedical Research Institute, Salt, Spain
| | - Dolors Capellà
- TransLab Research Group, and Department of Medical Sciences, University of Girona, Spain; Research Group on Statistics, Econometrics and Health, University of Girona, Spain; CIBER of Epidemiology and Public Health, Spain; Girona Biomedical Research Institute, Salt, Spain
| |
Collapse
|
26
|
Svoboda J, Popelikova A, Stuchlik A. Drugs Interfering with Muscarinic Acetylcholine Receptors and Their Effects on Place Navigation. Front Psychiatry 2017; 8:215. [PMID: 29170645 PMCID: PMC5684124 DOI: 10.3389/fpsyt.2017.00215] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 10/16/2017] [Indexed: 12/25/2022] Open
Abstract
Muscarinic acetylcholine receptors (mAChRs) have been found to regulate many diverse functions, ranging from motivation and feeding to spatial navigation, an important and widely studied type of cognitive behavior. Systemic administration of non-selective antagonists of mAChRs, such as scopolamine or atropine, have been found to have adverse effects on a vast majority of place navigation tasks. However, many of these results may be potentially confounded by disruptions of functions other than spatial learning and memory. Although studies with selective antimuscarinics point to mutually opposite effects of M1 and M2 receptors, their particular contribution to spatial cognition is still poorly understood, partly due to a lack of truly selective agents. Furthermore, constitutive knock-outs do not always support results from selective antagonists. For modeling impaired spatial cognition, the scopolamine-induced amnesia model still maintains some limited validity, but there is an apparent need for more targeted approaches such as local intracerebral administration of antagonists, as well as novel techniques such as optogenetics focused on cholinergic neurons and chemogenetics aimed at cells expressing metabotropic mAChRs.
Collapse
Affiliation(s)
- Jan Svoboda
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Anna Popelikova
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Ales Stuchlik
- Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
27
|
Abstract
Traditional antidepressant medications generally take weeks-to-months to achieve effect. However, the breakthrough finding of ketamine's rapidly acting antidepressant properties has inspired a decade-and-a-half of progress towards the identification of treatments that work quickly-within hours-to-days. This paradigm-shift in the discovery of antidepressant therapies has significantly changed the current landscape of antidepressant drug development. Building on this, the current review briefly highlights the recent trends in research towards identifying rapidly-acting antidepressants. Specifically, ketamine, GLYX-13, nitrous oxide, metabotropic glutamatergic receptor modulators, scopolamine, opioid-receptor modulators, and low field magnetic stimulation are discussed.
Collapse
|