1
|
Achtziger A, Alós-Ferrer C, Garagnani M. An inexpensive method to measure latent toxoplasmosis and its behavioral consequences. Acta Psychol (Amst) 2025; 254:104814. [PMID: 39999732 DOI: 10.1016/j.actpsy.2025.104814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/06/2024] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
An estimated 2.4 billion humans are affected by the protozoan Toxoplasma gondii, which causes one of the most common infections influencing human behavior and cognition. The infection and its effects are severely underresearched, because the diagnosis is costly and time-intensive, preventing widespread testing, in particular in less-than-wealthy regions. Psychological methods such as response time analysis could circumvent these problems and facilitate more intensive research. We propose a psychological diagnostic method for latent toxoplasmosis using response times, hence enabling large-scale tests of psychological, psychiatrical, and behavioral effects. A clinical trial (N = 119) showed that the method is sensitive (92 %) and specific (97 %) compared to testing for Toxoplasma IgG Antibodies (p < 0.0001, w = 0.85). A representative UK survey (N = 2020) using this method confirmed previously-hypothesized behavioral effects and uncovered previously-unknown effects of latent toxoplasmosis infections which are relevant and pervasive, both psychologically (increase in risky behaviors, depression, anxiety, and stress) and economically (decrease in yearly income of 2500 GBP; decrease in employment of 11 %).
Collapse
Affiliation(s)
- Anja Achtziger
- Department of Political and Social Sciences, Zeppelin University Friedrichshafen, Germany
| | | | - Michele Garagnani
- Centre for Brain, Mind and Markets, University of Melbourne, Melbourne, Australia
| |
Collapse
|
2
|
Bobkova NV, Chuvakova LN, Kovalev VI, Zhdanova DY, Chaplygina AV, Rezvykh AP, Evgen'ev MB. A Mouse Model of Sporadic Alzheimer's Disease with Elements of Major Depression. Mol Neurobiol 2025; 62:1337-1358. [PMID: 38980563 DOI: 10.1007/s12035-024-04346-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
After olfactory bulbectomy, animals are often used as a model of major depression or sporadic Alzheimer's disease and, hence, the status of this model is still disputable. To elucidate the nature of alterations in the expression of the genome after the operation, we analyzed transcriptomes of the cortex, hippocampus, and cerebellum of the olfactory bulbectomized (OBX) mice. Analysis of the functional significance of genes in the brain of OBX mice indicates that the balance of the GABA/glutamatergic systems is disturbed with hyperactivation of the latter in the hippocampus, leading to the development of excitotoxicity and induction of apoptosis in the background of severe mitochondrial dysfunction and astrogliosis. On top of this, the synthesis of neurotrophic factors decreases leading to the disruption of the cytoskeleton of neurons, an increase in the level of intracellular calcium, and the activation of tau protein hyperphosphorylation. Moreover, the acetylcholinergic system is deficient in the background of the hyperactivation of acetylcholinesterase. Importantly, the activity of the dopaminergic, endorphin, and opiate systems in OBX mice decreases, leading to hormonal dysfunction. On the other hand, genes responsible for the regulation of circadian rhythms, cell migration, and innate immunity are activated in OBX animals. All this takes place in the background of a drastic downregulation of ribosomal protein genes in the brain. The obtained results indicate that OBX mice represent a model of Alzheimer's disease with elements of major depression.
Collapse
Affiliation(s)
- N V Bobkova
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - L N Chuvakova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - V I Kovalev
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - D Y Zhdanova
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - A V Chaplygina
- Institute of Cell Biophysics of the Russian Academy of Sciences-Federal Research Center, Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - A P Rezvykh
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia
| | - M B Evgen'ev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991, Moscow, Russia.
| |
Collapse
|
3
|
Huang T, Hua Q, Zhao X, Tian W, Cao H, Xu W, Sun J, Zhang L, Wang K, Ji GJ. Abnormal functional lateralization and cooperation in bipolar disorder are associated with neurotransmitter and cellular profiles. J Affect Disord 2025; 369:970-977. [PMID: 39447972 DOI: 10.1016/j.jad.2024.10.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Hemispheric lateralization and cooperation are essential for efficient brain function, and aberrations in both have been found in psychiatric disorders such as schizophrenia. This study investigated alterations in hemispheric lateralization and cooperation among patients with bipolar disorder (BD) and associations with neurotransmitter and cell-type density distributions to identify potential molecular and cellular pathomechanisms. METHODS Sixty-seven BD patients and 127 healthy controls (HCs) were examined by resting-state functional MRI (rs-fMRI). Whole-brain maps of the autonomy index (AI) and connectivity between functionally homotopic voxels (CFH) were constructed to reveal BD-specific changes in brain functional lateralization and interhemispheric cooperation, respectively. Spatial associations of regional AI and CFH abnormalities with neurotransmitter and cell-type density distributions were examined by correlation analyses. RESULTS Bipolar disorder patients exhibited higher AI values in left superior parietal gyrus, cerebellar right Crus I, and cerebellar right Crus II, and these regional abnormalities were associated with the relative densities (proportions) of oligodendrocyte precursor cells and microglia. Patients also exhibited lower CFH values in right inferior parietal gyrus, bilateral middle occipital gyrus, left postcentral gyrus, and bilateral cerebellar crus II, and these regional abnormalities were associated with the densities of serotonin 1A and dopamine D2 receptors, oligodendrocyte precursor cells, astrocytes, and neurons. CONCLUSIONS These findings indicate that abnormal functional lateralization and cooperation in BD with potential molecular and cellular basis.
Collapse
Affiliation(s)
- Tongqing Huang
- School of Mental Health and Psychological Sciences, Anhui Medical University, 81 Meishan Rd, Hefei 230032, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Centre of Neuropsychiatric Disorder and Mental Health, Hefei, China
| | - Qiang Hua
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Centre of Neuropsychiatric Disorder and Mental Health, Hefei, China
| | - Xiya Zhao
- School of Mental Health and Psychological Sciences, Anhui Medical University, 81 Meishan Rd, Hefei 230032, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Centre of Neuropsychiatric Disorder and Mental Health, Hefei, China
| | - Weichao Tian
- School of Mental Health and Psychological Sciences, Anhui Medical University, 81 Meishan Rd, Hefei 230032, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Centre of Neuropsychiatric Disorder and Mental Health, Hefei, China
| | - Hai Cao
- School of Mental Health and Psychological Sciences, Anhui Medical University, 81 Meishan Rd, Hefei 230032, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Centre of Neuropsychiatric Disorder and Mental Health, Hefei, China
| | - Wenqiang Xu
- School of Mental Health and Psychological Sciences, Anhui Medical University, 81 Meishan Rd, Hefei 230032, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Centre of Neuropsychiatric Disorder and Mental Health, Hefei, China
| | - Jinmei Sun
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Centre of Neuropsychiatric Disorder and Mental Health, Hefei, China
| | - Li Zhang
- Department of Psychiatry, Affiliated Psychological Hospital of Anhui Medical University, Hefei, Anhui Province, China.
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Centre of Neuropsychiatric Disorder and Mental Health, Hefei, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China; Anhui Institute of Translational Medicine, Hefei, China.
| | - Gong-Jun Ji
- School of Mental Health and Psychological Sciences, Anhui Medical University, 81 Meishan Rd, Hefei 230032, China; Department of Psychology and Sleep Medicine, The Second Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei, China; Collaborative Innovation Centre of Neuropsychiatric Disorder and Mental Health, Hefei, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China; Anhui Institute of Translational Medicine, Hefei, China.
| |
Collapse
|
4
|
Bremshey S, Groß J, Renken K, Masseck OA. The role of serotonin in depression-A historical roundup and future directions. J Neurochem 2024; 168:1751-1779. [PMID: 38477031 DOI: 10.1111/jnc.16097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Depression is one of the most common psychiatric disorders worldwide, affecting approximately 280 million people, with probably much higher unrecorded cases. Depression is associated with symptoms such as anhedonia, feelings of hopelessness, sleep disturbances, and even suicidal thoughts. Tragically, more than 700 000 people commit suicide each year. Although depression has been studied for many decades, the exact mechanisms that lead to depression are still unknown, and available treatments only help a fraction of patients. In the late 1960s, the serotonin hypothesis was published, suggesting that serotonin is the key player in depressive disorders. However, this hypothesis is being increasingly doubted as there is evidence for the influence of other neurotransmitters, such as noradrenaline, glutamate, and dopamine, as well as larger systemic causes such as altered activity in the limbic network or inflammatory processes. In this narrative review, we aim to contribute to the ongoing debate on the involvement of serotonin in depression. We will review the evolution of antidepressant treatments, systemic research on depression over the years, and future research applications that will help to bridge the gap between systemic research and neurotransmitter dynamics using biosensors. These new tools in combination with systemic applications, will in the future provide a deeper understanding of the serotonergic dynamics in depression.
Collapse
Affiliation(s)
- Svenja Bremshey
- Synthetic Biology, University of Bremen, Bremen, Germany
- Neuropharmacology, University of Bremen, Bremen, Germany
| | - Juliana Groß
- Synthetic Biology, University of Bremen, Bremen, Germany
| | - Kim Renken
- Synthetic Biology, University of Bremen, Bremen, Germany
| | | |
Collapse
|
5
|
Harkin EF, Nasrallah G, Le François B, Albert PR. Transcriptional Regulation of the Human 5-HT1A Receptor Gene by Lithium: Role of Deaf1 and GSK3β. Int J Mol Sci 2023; 24:15620. [PMID: 37958600 PMCID: PMC10647674 DOI: 10.3390/ijms242115620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/11/2023] [Accepted: 10/21/2023] [Indexed: 11/15/2023] Open
Abstract
Serotonin 1A (5-HT1A) autoreceptors located on serotonin neurons inhibit their activity, and their upregulation has been implicated in depression, suicide and resistance to antidepressant treatment. Conversely, post-synaptic 5-HT1A heteroreceptors are important for antidepressant response. The transcription factor deformed epidermal autoregulatory factor 1 (Deaf1) acts as a presynaptic repressor and postsynaptic enhancer of 5-HT1A transcription, but the mechanism is unclear. Because Deaf1 interacts with and is phosphorylated by glycogen synthase kinase 3β (GSK3β)-a constitutively active protein kinase that is inhibited by the mood stabilizer lithium at therapeutic concentrations-we investigated the role of GSK3β in Deaf1 regulation of human 5-HT1A transcription. In 5-HT1A promoter-reporter assays, human HEK293 kidney and 5-HT1A-expressing SKN-SH neuroblastoma cells, transfection of Deaf1 reduced 5-HT1A promoter activity by ~45%. To identify potential GSK3β site(s) on Deaf1, point mutations of known and predicted phosphorylation sites on Deaf1 were tested. Deaf1 repressor function was not affected by any of the mutants tested except the Y300F mutant, which augmented Deaf1 repression. Both lithium and the selective GSK3 inhibitors CHIR-99021 and AR-014418 attenuated and reversed Deaf1 repression compared to vector. This inhibition was at concentrations that maximally inhibit GSK3β activity as detected by the GSK3β-sensitive TCF/LEF reporter construct. Our results support the hypothesis that GSK3β regulates the activity of Deaf1 to repress 5-HT1A transcription and provide a potential mechanism for actions of GSK3 inhibitors on behavior.
Collapse
Affiliation(s)
| | | | | | - Paul R. Albert
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa, 451 Smyth Road, Ottawa, ON K1H-8M5, Canada (B.L.F.)
| |
Collapse
|
6
|
Galfalvy H, Shea E, de Vegvar J, Pantazatos S, Huang YY, Burke AK, Sublette ME, Oquendo MA, Zanderigo F, Miller JM, Mann JJ. Brain serotonin 1A receptor binding: relationship to peripheral blood DNA methylation, recent life stress and childhood adversity in unmedicated major depression. Br J Psychiatry 2023; 223:415-421. [PMID: 37395098 PMCID: PMC10514224 DOI: 10.1192/bjp.2023.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 12/19/2022] [Accepted: 01/12/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Childhood and lifetime adversity may reduce brain serotonergic (5-HT) neurotransmission by epigenetic mechanisms. AIMS We tested the relationships of childhood adversity and recent stress to serotonin 1A (5-HT1A) receptor genotype, DNA methylation of this gene in peripheral blood monocytes and in vivo 5-HT1A receptor binding potential (BPF) determined by positron emission tomography (PET) in 13 a priori brain regions, in participants with major depressive disorder (MDD) and healthy volunteers (controls). METHOD Medication-free participants with MDD (n = 192: 110 female, 81 male, 1 other) and controls (n = 88: 48 female, 40 male) were interviewed about childhood adversity and recent stressors and genotyped for rs6295. DNA methylation was assayed at three upstream promoter sites (-1019, -1007, -681) of the 5-HT1A receptor gene. A subgroup (n = 119) had regional brain 5-HT1A receptor BPF quantified by PET. Multi-predictor models were used to test associations between diagnosis, recent stress, childhood adversity, genotype, methylation and BPF. RESULTS Recent stress correlated positively with blood monocyte methylation at the -681 CpG site, adjusted for diagnosis, and had positive and region-specific correlations with 5-HT1A BPF in participants with MDD, but not in controls. In participants with MDD, but not in controls, methylation at the -1007 CpG site had positive and region-specific correlations with binding potential. Childhood adversity was not associated with methylation or BPF in participants with MDD. CONCLUSIONS These findings support a model in which recent stress increases 5-HT1A receptor binding, via methylation of promoter sites, thus affecting MDD psychopathology.
Collapse
MESH Headings
- Humans
- Male
- Female
- Depressive Disorder, Major/diagnostic imaging
- Depressive Disorder, Major/genetics
- Depressive Disorder, Major/drug therapy
- Receptor, Serotonin, 5-HT1A/genetics
- Receptor, Serotonin, 5-HT1A/metabolism
- Receptor, Serotonin, 5-HT1A/therapeutic use
- DNA Methylation
- Serotonin/metabolism
- Serotonin/therapeutic use
- Depression
- Brain/pathology
- Positron-Emission Tomography/methods
- Stress, Psychological/genetics
Collapse
Affiliation(s)
- Hanga Galfalvy
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Department of Biostatistics, Columbia University Mailman School of Public Health, New York, New York, USA
| | | | - Jacqueline de Vegvar
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Spiro Pantazatos
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA
| | - Yung-yu Huang
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA
| | - Ainsley K. Burke
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA
| | - M. Elizabeth Sublette
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA
| | - Maria A. Oquendo
- Psychiatry Department, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Francesca Zanderigo
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA
| | - Jeffrey M. Miller
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; and Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA
| | - J. John Mann
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA; Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, New York, USA; and Department of Radiology, Columbia University, New York, New York, USA
| |
Collapse
|
7
|
Khani P, Ansari Dezfouli M, Nasri F, Rahemi M, Ahmadloo S, Afkhami H, Saeidi F, Tereshchenko S, Bigdeli MR, Modarressi MH. Genetic and epigenetic effects on couple adjustment in context of romantic relationship: A scoping systematic review. Front Genet 2023; 14:1002048. [PMID: 36816018 PMCID: PMC9937082 DOI: 10.3389/fgene.2023.1002048] [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] [Received: 07/24/2022] [Accepted: 01/02/2023] [Indexed: 01/26/2023] Open
Abstract
Introduction: Couples' relationships defined by a complex interaction between the two partners and their intrapersonal traits. Romantic; relationships and love are associated with marital satisfaction and stability, as well as couples' happiness and health. Personality traits influence romantic relationships and, personality influenced by genetical and non-genetically factors. The roles of non-genetically factors such as socioeconomic position and external appearance have revealed in determining the quality of romantic relationships. Methods: We; performed a scoping systematic review to assess the association between genetics and epigenetic factors and romantic relationship. Relevant articles were identified by PubMed, EMBASE, Web of Science, Scopus, and the APA PsycInfo searching between inception and 4 June 2022. Results: Different studies evaluated the associated polymorphisms in 15 different genes or chromosomal regions. In the first step; we classified them into four groups: (1) Oxytocin-related signaling pathway (OXTR, CD38, and AVPR1A); (2) Serotonin-related signaling pathway (SLC6A4, HTR1A, and HTR2A); (3) Dopamine and catecholamine-related signaling pathway (DRD1, DRD2, DRD4, ANKK1, and COMT); and (4) other genes (HLA, GABRA2, OPRM1, and Y-DNA haplogroup D-M55). Then, we evaluated and extracted significant polymorphisms that affect couple adjustment and romantic relationships. Discussion: Overall, the findings suggest that genetic and epigenetics variants play a key role in marital adjustment and romantic relationships over time.
Collapse
Affiliation(s)
- Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mitra Ansari Dezfouli
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Farzad Nasri
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Rahemi
- Department of stem cell technology and tissue regeneration, Faculty of Science, Tehran University, Tehran, Iran
| | - Salma Ahmadloo
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Institute for Cognitive and Brain Sciences, Shahid Beheshti University, Tehran, Iran
| | - Hamed Afkhami
- Department of Medical Microbiology, Faculty of Medicine, Shahed University of Medical Sciences, Tehran, Iran
| | - Farzane Saeidi
- Department of Medical Genetics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sergey Tereshchenko
- Research Institute of Medical Problems of the North, Federal Research Center “Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences”, Krasnoyarsk, Russia
| | - Mohammad Reza Bigdeli
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Institute for Cognitive and Brain Sciences, Shahid Beheshti University, Tehran, Iran
| | - Mohammad Hossein Modarressi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
8
|
Simayi D, Guan Y. HTR1A Gene Polymorphism in Type 2 Diabetes Mellitus Comorbid with Major Depressive Disorder in a Chinese Population. Diabetes Metab Syndr Obes 2022; 15:1597-1604. [PMID: 35651901 PMCID: PMC9148918 DOI: 10.2147/dmso.s361843] [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: 02/25/2022] [Accepted: 05/11/2022] [Indexed: 04/20/2023] Open
Abstract
Background Major depressive disorder is a frequent mental illness, which is common in patients with type 2 diabetes. Type 2 diabetes comorbid with depression has a worse prognosis. There are multiple risk factors for depression, and genetic studies have shown that gene polymorphism may play an important role in the pathogenesis of depression. Methods A total of 874 patients with type 2 diabetes were recruited for this study and divided into two groups: depressive group (DDM group, n = 234) and non-depressive group (NDDM group, n = 640). HTR1A gene polymorphisms (rs6295, rs878567, rs1800044) genotyping work was performed using a custom by design 48-Plex SNPscanTM Kit. Results The rs6295, rs878567, and rs1800044 SNPs were not associated with type 2 diabetes comorbid with depression. Female sex, age, and FBG level increased the risk of depression in patients with type 2 diabetes. Conclusion HTR1A rs6295, rs878567, and rs1800044 SNPs polymorphism is not associated with type 2 diabetes comorbid with depression. Rather, female sex, age, and FBG level are risk factors for depression among patients with type 2 diabetes. Larger studies are needed to further confirm our findings.
Collapse
Affiliation(s)
- Dilixia Simayi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
- Department of Oncology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| | - Yaqun Guan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, Xinjiang, People’s Republic of China
| |
Collapse
|
9
|
Kuhns L, Kroon E, Colyer-Patel K, Cousijn J. Associations between cannabis use, cannabis use disorder, and mood disorders: longitudinal, genetic, and neurocognitive evidence. Psychopharmacology (Berl) 2022; 239:1231-1249. [PMID: 34741634 PMCID: PMC9520129 DOI: 10.1007/s00213-021-06001-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 10/11/2021] [Indexed: 12/16/2022]
Abstract
RATIONALE Cannabis use among people with mood disorders increased in recent years. While comorbidity between cannabis use, cannabis use disorder (CUD), and mood disorders is high, the underlying mechanisms remain unclear. OBJECTIVES We aimed to evaluate (1) the epidemiological evidence for an association between cannabis use, CUD, and mood disorders; (2) prospective longitudinal, genetic, and neurocognitive evidence of underlying mechanisms; and (3) prognosis and treatment options for individuals with CUD and mood disorders. METHODS Narrative review of existing literature is identified through PubMed searches, reviews, and meta-analyses. Evidence was reviewed separately for depression, bipolar disorder, and suicide. RESULTS Current evidence is limited and mixed but suggestive of a bidirectional relationship between cannabis use, CUD, and the onset of depression. The evidence more consistently points to cannabis use preceding onset of bipolar disorder. Shared neurocognitive mechanisms and underlying genetic and environmental risk factors appear to explain part of the association. However, cannabis use itself may also influence the development of mood disorders, while others may initiate cannabis use to self-medicate symptoms. Comorbid cannabis use and CUD are associated with worse prognosis for depression and bipolar disorder including increased suicidal behaviors. Evidence for targeted treatments is limited. CONCLUSIONS The current evidence base is limited by the lack of well-controlled prospective longitudinal studies and clinical studies including comorbid individuals. Future studies in humans examining the causal pathways and potential mechanisms of the association between cannabis use, CUD, and mood disorder comorbidity are crucial for optimizing harm reduction and treatment strategies.
Collapse
Affiliation(s)
- Lauren Kuhns
- Department of Psychology, Neuroscience of Addiction (NofA, University of Amsterdam, Amsterdam, the Netherlands.
- The Amsterdam Brain and Cognition Center (ABC), University of Amsterdam, Amsterdam, the Netherlands.
| | - Emese Kroon
- Department of Psychology, Neuroscience of Addiction (NofA, University of Amsterdam, Amsterdam, the Netherlands
- The Amsterdam Brain and Cognition Center (ABC), University of Amsterdam, Amsterdam, the Netherlands
| | - Karis Colyer-Patel
- Department of Psychology, Neuroscience of Addiction (NofA, University of Amsterdam, Amsterdam, the Netherlands
| | - Janna Cousijn
- Department of Psychology, Neuroscience of Addiction (NofA, University of Amsterdam, Amsterdam, the Netherlands
- The Amsterdam Brain and Cognition Center (ABC), University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
10
|
Anguita-Ruiz A, Zarza-Rebollo JA, Pérez-Gutiérrez AM, Molina E, Gutiérrez B, Bellón JÁ, Moreno-Peral P, Conejo-Cerón S, Aiarzagüena JM, Ballesta-Rodríguez MI, Fernández A, Fernández-Alonso C, Martín-Pérez C, Montón-Franco C, Rodríguez-Bayón A, Torres-Martos Á, López-Isac E, Cervilla J, Rivera M. Body mass index interacts with a genetic-risk score for depression increasing the risk of the disease in high-susceptibility individuals. Transl Psychiatry 2022; 12:30. [PMID: 35075110 PMCID: PMC8786870 DOI: 10.1038/s41398-022-01783-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 11/24/2021] [Accepted: 01/04/2022] [Indexed: 11/22/2022] Open
Abstract
Depression is strongly associated with obesity among other chronic physical diseases. The latest mega- and meta-analysis of genome-wide association studies have identified multiple risk loci robustly associated with depression. In this study, we aimed to investigate whether a genetic-risk score (GRS) combining multiple depression risk single nucleotide polymorphisms (SNPs) might have utility in the prediction of this disorder in individuals with obesity. A total of 30 depression-associated SNPs were included in a GRS to predict the risk of depression in a large case-control sample from the Spanish PredictD-CCRT study, a national multicentre, randomized controlled trial, which included 104 cases of depression and 1546 controls. An unweighted GRS was calculated as a summation of the number of risk alleles for depression and incorporated into several logistic regression models with depression status as the main outcome. Constructed models were trained and evaluated in the whole recruited sample. Non-genetic-risk factors were combined with the GRS in several ways across the five predictive models in order to improve predictive ability. An enrichment functional analysis was finally conducted with the aim of providing a general understanding of the biological pathways mapped by analyzed SNPs. We found that an unweighted GRS based on 30 risk loci was significantly associated with a higher risk of depression. Although the GRS itself explained a small amount of variance of depression, we found a significant improvement in the prediction of depression after including some non-genetic-risk factors into the models. The highest predictive ability for depression was achieved when the model included an interaction term between the GRS and the body mass index (BMI), apart from the inclusion of classical demographic information as marginal terms (AUC = 0.71, 95% CI = [0.65, 0.76]). Functional analyses on the 30 SNPs composing the GRS revealed an over-representation of the mapped genes in signaling pathways involved in processes such as extracellular remodeling, proinflammatory regulatory mechanisms, and circadian rhythm alterations. Although the GRS on its own explained a small amount of variance of depression, a significant novel feature of this study is that including non-genetic-risk factors such as BMI together with a GRS came close to the conventional threshold for clinical utility used in ROC analysis and improves the prediction of depression. In this study, the highest predictive ability was achieved by the model combining the GRS and the BMI under an interaction term. Particularly, BMI was identified as a trigger-like risk factor for depression acting in a concerted way with the GRS component. This is an interesting finding since it suggests the existence of a risk overlap between both diseases, and the need for individual depression genetics-risk evaluation in subjects with obesity. This research has therefore potential clinical implications and set the basis for future research directions in exploring the link between depression and obesity-associated disorders. While it is likely that future genome-wide studies with large samples will detect novel genetic variants associated with depression, it seems clear that a combination of genetics and non-genetic information (such is the case of obesity status and other depression comorbidities) will still be needed for the optimization prediction of depression in high-susceptibility individuals.
Collapse
Affiliation(s)
- Augusto Anguita-Ruiz
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain ,grid.4489.10000000121678994Institute of Nutrition and Food Technology “José Mataix”, Biomedical Research Center (CIBM), University of Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.413448.e0000 0000 9314 1427CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Juan Antonio Zarza-Rebollo
- Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain. .,Institute of Neurosciences 'Federico Olóriz', Biomedical Research Center (CIBM), University of Granada, Granada, Spain.
| | - Ana M Pérez-Gutiérrez
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain ,grid.4489.10000000121678994Institute of Neurosciences ‘Federico Olóriz’, Biomedical Research Center (CIBM), University of Granada, Granada, Spain
| | - Esther Molina
- grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.4489.10000000121678994Institute of Neurosciences ‘Federico Olóriz’, Biomedical Research Center (CIBM), University of Granada, Granada, Spain ,grid.4489.10000000121678994Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain
| | - Blanca Gutiérrez
- grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.4489.10000000121678994Institute of Neurosciences ‘Federico Olóriz’, Biomedical Research Center (CIBM), University of Granada, Granada, Spain ,grid.4489.10000000121678994Department of Psychiatry, Faculty of Medicine, University of Granada, Granada, Spain
| | - Juan Ángel Bellón
- grid.452525.1Primary Care District of Málaga-Guadalhorce, Biomedical Research Institute of Málaga (IBIMA), Primary Care Prevention and Health Promotion Network (redIAPP), Málaga, Spain ,grid.10215.370000 0001 2298 7828Department of Public Health and Psychiatry, Faculty of Medicine, University of Málaga, Málaga, Spain
| | - Patricia Moreno-Peral
- grid.452525.1Primary Care District of Málaga-Guadalhorce, Biomedical Research Institute of Málaga (IBIMA), Primary Care Prevention and Health Promotion Network (redIAPP), Málaga, Spain
| | - Sonia Conejo-Cerón
- grid.452525.1Primary Care District of Málaga-Guadalhorce, Biomedical Research Institute of Málaga (IBIMA), Primary Care Prevention and Health Promotion Network (redIAPP), Málaga, Spain
| | | | | | - Anna Fernández
- grid.428876.7Parc Sanitari Sant Joan de Déu, Fundació Sant Joan de Déu, Barcelona, Spain ,grid.466571.70000 0004 1756 6246CIBERESP, Centro de Investigacion Biomedica en Red de Epidemiologia y Salud Publica, Madrid, Spain
| | | | - Carlos Martín-Pérez
- grid.418355.eMarquesado Health Centre, Servicio Andaluz de Salud, Granada, Spain
| | - Carmen Montón-Franco
- grid.488737.70000000463436020Casablanca Health Centre, Aragonese Institute of Health Sciences, IIS Aragón, Zaragoza, Spain ,grid.11205.370000 0001 2152 8769Department of Medicine and Psychiatry, University of Zaragoza, Zaragoza, Spain
| | | | - Álvaro Torres-Martos
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Elena López-Isac
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain ,grid.4489.10000000121678994Institute of Neurosciences ‘Federico Olóriz’, Biomedical Research Center (CIBM), University of Granada, Granada, Spain
| | - Jorge Cervilla
- grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.4489.10000000121678994Institute of Neurosciences ‘Federico Olóriz’, Biomedical Research Center (CIBM), University of Granada, Granada, Spain ,grid.4489.10000000121678994Department of Psychiatry, Faculty of Medicine, University of Granada, Granada, Spain
| | - Margarita Rivera
- grid.4489.10000000121678994Department of Biochemistry and Molecular Biology II, Faculty of Pharmacy, University of Granada, Granada, Spain ,grid.507088.2Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain ,grid.4489.10000000121678994Institute of Neurosciences ‘Federico Olóriz’, Biomedical Research Center (CIBM), University of Granada, Granada, Spain
| |
Collapse
|
11
|
Terock J, Weihs A, Teumer A, Klinger-König J, Janowitz D, Grabe HJ. Associations and interactions of the serotonin receptor genes 5-HT1A, 5-HT2A, and childhood trauma with alexithymia in two independent general-population samples. Psychiatry Res 2021; 298:113783. [PMID: 33567384 DOI: 10.1016/j.psychres.2021.113783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/31/2021] [Indexed: 12/18/2022]
Abstract
Previous studies suggested that childhood trauma and a disturbed serotonergic neurotransmission are involved in the pathogenesis of alexithymia. Specifically, genetic polymorphisms of the serotonin receptors 5-HT1A and 5-HT2A were found to be associated with alexithymia. However, it is unclear whether these factors show main or interaction effects with childhood trauma on alexithymia. Data from two independent general-population cohorts of the Study of Health in Pomerania (SHIP-Trend: N=3,706, Age: range=20-83, 51.6% female, SHIP-LEGEND: N=2,162, Age: range=20-80, 52.5% female) were used. The Toronto Alexithymia Scale-20 (TAS-20) and the Childhood Trauma Questionnaire (CTQ) were applied. Genotypes of rs6295 of 5-HT1A and rs6311 of 5-HT2A were determined. Ordinary least-squared regression models with robust standard errors were applied to investigate associations of the main and interaction effects of childhood maltreatment and the polymorphisms with alexithymia. Childhood trauma, but none of the investigated polymorphisms showed main effects on alexithymia. However, childhood trauma showed significant CTQ sum score x rs6295 interactions in male subjects in both samples such that the presence of the G-allele diminished the CTQ associated increase in the TAS-20 sum scores. Our results support a strong role of early life stress and interactions with rs6295 on alexithymic personality features at least in male subjects.
Collapse
Affiliation(s)
- Jan Terock
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany; Department of Psychiatry and Psychotherapy, HELIOS Hanseklinikum Stralsund, Stralsund, Germany.
| | - Antoine Weihs
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Johanna Klinger-König
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Deborah Janowitz
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany; German Center for Neurodegenerative Diseases DZNE, Site Rostock/ Greifswald, Germany
| |
Collapse
|
12
|
Jo K, Kim S, Hong KB, Suh HJ. Nelumbo nucifera promotes non-rapid eye movement sleep by regulating GABAergic receptors in rat model. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113511. [PMID: 33148434 DOI: 10.1016/j.jep.2020.113511] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/21/2020] [Accepted: 10/20/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Nelumbo nucifera are used in folk medicine for anti-depressant, anti-convulsant, neuroprotective, and many other purposes. AIM OF THE STUDY The present work evaluated the sleep potentiating effects of water extract from lotus seed in rat, and the neuropharmacological mechanisms underlying these effects. MATERIALS AND METHODS Pentobarbital-induced sleep test and electroencephalogram (EEG) analysis were applied to investigate sleep latency, duration, total sleeping time and sleep quality of Lotus extract. In addition, real-time PCR and HPLC analysis were applied to analyze the signaling pathway. RESULTS We found that the amounts of the possible active compounds GABA (2.33 mg/g) and L-tryptophan (2.00 mg/g) were higher than quinidine (0.55 mg/g) and neferine (0.16 mg/g) in lotus seed extract. High dose (160 mg/kg) administration of lotus extract led to a tendency towards decreased sleep latency time and an increase in sleep duration time compared to the control group in a pentobarbital-induced sleep model (p < 0.05). After high dose administration, total sleep and NREM were significantly increased compared to control, while wake time and REM were significantly decreased. Lotus extract-treated rats showed significantly reduced wake time and increased sleep time in a caffeine-induced model of arousal. The transcription level of GABAA receptor, GABAB receptor, and serotonin receptor tended to increase with dose, and lotus extract showed a strong dose-dependent binding capacity to the GABAA receptor. CONCLUSION The above results strongly suggest that GABA contained in lotus seed extract acts as a sleep potentiating compound, and that sleep-potentiating activity involves GABAA receptor binding.
Collapse
Affiliation(s)
- Kyungae Jo
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea Univetsity, Seoul, 02841, Republic of Korea.
| | - Singeun Kim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea Univetsity, Seoul, 02841, Republic of Korea.
| | - Ki-Bae Hong
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea Univetsity, Seoul, 02841, Republic of Korea.
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea Univetsity, Seoul, 02841, Republic of Korea.
| |
Collapse
|
13
|
Buch AM, Liston C. Dissecting diagnostic heterogeneity in depression by integrating neuroimaging and genetics. Neuropsychopharmacology 2021; 46:156-175. [PMID: 32781460 PMCID: PMC7688954 DOI: 10.1038/s41386-020-00789-3] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 07/07/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022]
Abstract
Depression is a heterogeneous and etiologically complex psychiatric syndrome, not a unitary disease entity, encompassing a broad spectrum of psychopathology arising from distinct pathophysiological mechanisms. Motivated by a need to advance our understanding of these mechanisms and develop new treatment strategies, there is a renewed interest in investigating the neurobiological basis of heterogeneity in depression and rethinking our approach to diagnosis for research purposes. Large-scale genome-wide association studies have now identified multiple genetic risk variants implicating excitatory neurotransmission and synapse function and underscoring a highly polygenic inheritance pattern that may be another important contributor to heterogeneity in depression. Here, we review various sources of phenotypic heterogeneity and approaches to defining and studying depression subtypes, including symptom-based subtypes and biology-based approaches to decomposing the depression syndrome. We review "dimensional," "categorical," and "hybrid" approaches to parsing phenotypic heterogeneity in depression and defining subtypes using functional neuroimaging. Next, we review recent progress in neuroimaging genetics (correlating neuroimaging patterns of brain function with genetic data) and its potential utility for generating testable hypotheses concerning molecular and circuit-level mechanisms. We discuss how genetic variants and transcriptomic profiles may confer risk for depression by modulating brain structure and function. We conclude by highlighting several promising areas for future research into the neurobiological underpinnings of heterogeneity, including efforts to understand sexually dimorphic mechanisms, the longitudinal dynamics of depressive episodes, and strategies for developing personalized treatments and facilitating clinical decision-making.
Collapse
Affiliation(s)
- Amanda M Buch
- Department of Psychiatry and Brain and Mind Research Institute, Weill Cornell Medicine, 413 East 69th Street, Box 240, New York, NY, 10021, USA
| | - Conor Liston
- Department of Psychiatry and Brain and Mind Research Institute, Weill Cornell Medicine, 413 East 69th Street, Box 240, New York, NY, 10021, USA.
| |
Collapse
|
14
|
Toxoplasma gondii: AnUnderestimated Threat? Trends Parasitol 2020; 36:959-969. [PMID: 33012669 DOI: 10.1016/j.pt.2020.08.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/17/2020] [Accepted: 08/21/2020] [Indexed: 02/08/2023]
Abstract
Traditionally, the protozoan parasite Toxoplasma gondii has been thought of as relevant to public health primarily within the context of congenital toxoplasmosis or postnatally acquired disease in immunocompromised patients. However, latent T.gondii infection has been increasingly associated with a wide variety of neuropsychiatric disorders and, more recently, causal frameworks for these epidemiological associations have been proposed. We present assimilated evidence on the associations between T.gondii and various human neuropsychiatric disorders and outline how these may be explained within a unifying causal framework. We argue that the occult effects of latent T.gondii infection likely outweigh the recognised overt morbidity caused by toxoplasmosis, substantially raising the public health importance of this parasite.
Collapse
|
15
|
Li Y, Xiao H, Dong J, Luo D, Wang H, Zhang S, Zhu T, Zhu C, Cui M, Fan S. Gut Microbiota Metabolite Fights Against Dietary Polysorbate 80-Aggravated Radiation Enteritis. Front Microbiol 2020; 11:1450. [PMID: 32670255 PMCID: PMC7332576 DOI: 10.3389/fmicb.2020.01450] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
Radiation therapy is a cornerstone of modern management methods for malignancies but is accompanied by diverse side effects. In the present study, we showed that food additives such as polysorbate 80 (P80) exacerbate irradiation-induced gastrointestinal (GI) tract toxicity. A 16S ribosomal RNA high-throughput sequencing analysis indicated that P80 consumption altered the abundance and composition of the gut microbiota, leading to severe radiation-induced GI tract injury. Mice harboring fecal microbes from P80-treated mice were highly susceptible to irradiation, and antibiotics-challenged mice also represented more sensitive to radiation following P80 treatment. Importantly, butyrate, a major metabolite of enteric microbial fermentation of dietary fibers, exhibited beneficial effects against P80 consumption-aggravated intestinal toxicity via the activation of G-protein-coupled receptors (GPCRs) and maintenance of the intestinal bacterial composition in irradiated animals. Moreover, butyrate had broad therapeutic effects on common radiation-induced injury. Collectively, our findings demonstrate that P80 are potential risk factors for cancer patients during radiotherapy and indicate that butyrate might be employed as a therapeutic option to mitigate the complications associated with radiotherapy.
Collapse
Affiliation(s)
- Yuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Huiwen Xiao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiali Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Dan Luo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Haichao Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China.,Department of Emergency Medicine, North Shore University Hospital, Manhasset, NY, United States.,Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Shuqin Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Tong Zhu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Changchun Zhu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
16
|
Maul S, Giegling I, Fabbri C, Corponi F, Serretti A, Rujescu D. Genetics of resilience: Implications from genome-wide association studies and candidate genes of the stress response system in posttraumatic stress disorder and depression. Am J Med Genet B Neuropsychiatr Genet 2020; 183:77-94. [PMID: 31583809 DOI: 10.1002/ajmg.b.32763] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/22/2019] [Accepted: 09/03/2019] [Indexed: 12/28/2022]
Abstract
Resilience is the ability to cope with critical situations through the use of personal and socially mediated resources. Since a lack of resilience increases the risk of developing stress-related psychiatric disorders such as posttraumatic stress disorder (PTSD) and major depressive disorder (MDD), a better understanding of the biological background is of great value to provide better prevention and treatment options. Resilience is undeniably influenced by genetic factors, but very little is known about the exact underlying mechanisms. A recently published genome-wide association study (GWAS) on resilience has identified three new susceptibility loci, DCLK2, KLHL36, and SLC15A5. Further interesting results can be found in association analyses of gene variants of the stress response system, which is closely related to resilience, and PTSD and MDD. Several promising genes, such as the COMT (catechol-O-methyltransferase) gene, the serotonin transporter gene (SLC6A4), and neuropeptide Y (NPY) suggest gene × environment interaction between genetic variants, childhood adversity, and the occurrence of PTSD and MDD, indicating an impact of these genes on resilience. GWAS on PTSD and MDD provide another approach to identifying new disease-associated loci and, although the functional significance for disease development for most of these risk genes is still unknown, they are potential candidates due to the overlap of stress-related psychiatric disorders and resilience. In the future, it will be important for genetic studies to focus more on resilience than on pathological phenotypes, to develop reasonable concepts for measuring resilience, and to establish international cooperations to generate sufficiently large samples.
Collapse
Affiliation(s)
- Stephan Maul
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Ina Giegling
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Chiara Fabbri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Filippo Corponi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Dan Rujescu
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
17
|
Fortinguerra S, Sorrenti V, Giusti P, Zusso M, Buriani A. Pharmacogenomic Characterization in Bipolar Spectrum Disorders. Pharmaceutics 2019; 12:E13. [PMID: 31877761 PMCID: PMC7022469 DOI: 10.3390/pharmaceutics12010013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/14/2019] [Accepted: 12/19/2019] [Indexed: 12/15/2022] Open
Abstract
The holistic approach of personalized medicine, merging clinical and molecular characteristics to tailor the diagnostic and therapeutic path to each individual, is steadily spreading in clinical practice. Psychiatric disorders represent one of the most difficult diagnostic challenges, given their frequent mixed nature and intrinsic variability, as in bipolar disorders and depression. Patients misdiagnosed as depressed are often initially prescribed serotonergic antidepressants, a treatment that can exacerbate a previously unrecognized bipolar condition. Thanks to the use of the patient's genomic profile, it is possible to recognize such risk and at the same time characterize specific genetic assets specifically associated with bipolar spectrum disorder, as well as with the individual response to the various therapeutic options. This provides the basis for molecular diagnosis and the definition of pharmacogenomic profiles, thus guiding therapeutic choices and allowing a safer and more effective use of psychotropic drugs. Here, we report the pharmacogenomics state of the art in bipolar disorders and suggest an algorithm for therapeutic regimen choice.
Collapse
Affiliation(s)
- Stefano Fortinguerra
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group (Synlab Limited), 35131 Padova, Italy; (S.F.); (V.S.)
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (P.G.); (M.Z.)
| | - Vincenzo Sorrenti
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group (Synlab Limited), 35131 Padova, Italy; (S.F.); (V.S.)
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (P.G.); (M.Z.)
- Bendessere™ Study Center, Solgar Italia Multinutrient S.p.A., 35131 Padova, Italy
| | - Pietro Giusti
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (P.G.); (M.Z.)
| | - Morena Zusso
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (P.G.); (M.Z.)
| | - Alessandro Buriani
- Maria Paola Belloni Center for Personalized Medicine, Data Medica Group (Synlab Limited), 35131 Padova, Italy; (S.F.); (V.S.)
- Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131 Padova, Italy; (P.G.); (M.Z.)
| |
Collapse
|
18
|
Brunoni AR, Carracedo A, Amigo OM, Pellicer AL, Talib L, Carvalho AF, Lotufo PA, Benseñor IM, Gattaz W, Cappi C. Association of BDNF, HTR2A, TPH1, SLC6A4, and COMT polymorphisms with tDCS and escitalopram efficacy: ancillary analysis of a double-blind, placebo-controlled trial. ACTA ACUST UNITED AC 2019; 42:128-135. [PMID: 31721892 PMCID: PMC7115450 DOI: 10.1590/1516-4446-2019-0620] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 08/20/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVE We investigated whether single nucleotide polymorphisms (SNPs) associated with neuroplasticity and activity of monoamine neurotransmitters, such as the brain-derived neurotrophic factor (BDNF, rs6265), the serotonin transporter (SLC6A4, rs25531), the tryptophan hydroxylase 1 (TPH1, rs1800532), the 5-hydroxytryptamine receptor 2A (HTR2A, rs6311, rs6313, rs7997012), and the catechol-O-methyltransferase (COMT, rs4680) genes, are associated with efficacy of transcranial direct current stimulation (tDCS) in major depression. METHODS Data from the Escitalopram vs. Electrical Current Therapy for Treating Depression Clinical Study (ELECT-TDCS) were used. Participants were antidepressant-free at baseline and presented with an acute, moderate-to-severe unipolar depressive episode. They were randomized to receive escitalopram/tDCS-sham (n=75), tDCS/placebo-pill (n=75), or placebo-pill/sham-tDCS (n=45). General linear models assessed the interaction between treatment group and allele-wise carriers. Additional analyses were performed for each group and each genotype separately. RESULTS Pairwise group comparisons (tDCS vs. placebo, tDCS vs. escitalopram, and escitalopram vs. placebo) did not identify alleles associated with depression improvement. In addition, exploratory analyses also did not identify any SNP unequivocally associated with improvement of depression in any treatment group. CONCLUSION Larger, combined datasets are necessary to identify candidate genes for tDCS response.
Collapse
Affiliation(s)
- Andre R Brunoni
- Departamento de Medicina Interna, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Angel Carracedo
- Grupo de Medicina Xenómica/Pharmacogenetics Research, Laboratorio SSL1, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Santiago de Compostela, Spain
| | - Olalla M Amigo
- Grupo de Medicina Xenómica/Pharmacogenetics Research, Laboratorio SSL1, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Santiago de Compostela, Spain
| | - Ana L Pellicer
- Grupo de Medicina Xenómica/Pharmacogenetics Research, Laboratorio SSL1, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CiMUS), Santiago de Compostela, Spain
| | - Leda Talib
- Laboratório de Neurociências (LIM-27) and Instituto Nacional de Biomarcadores em Psiquiatria (INBION), Departamento e Instituto de Psiquiatria, Faculdade de Medicina, São Paulo, SP, Brazil
| | - Andre F Carvalho
- Department of Psychiatry, Faculty of Medicine, University of Toronto & Centre for Addiction & Mental Health (CAMH), Toronto, Canada
| | - Paulo A Lotufo
- Departamento de Medicina Interna, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Isabela M Benseñor
- Departamento de Medicina Interna, Faculdade de Medicina, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Wagner Gattaz
- Laboratório de Neurociências (LIM-27) and Instituto Nacional de Biomarcadores em Psiquiatria (INBION), Departamento e Instituto de Psiquiatria, Faculdade de Medicina, São Paulo, SP, Brazil
| | - Carolina Cappi
- Programa Transtornos do Espectro Obsessivo-Compulsivo, Departamento e Instituto de Psiquiatria, Faculdade de Medicina, USP, São Paulo, SP, Brazil
| |
Collapse
|
19
|
Davydova YD, Enikeeva RF, Kazantseva AV, Mustafin RN, Romanova AR, Malykh SB, Khusnutdinov EK. Genetic basis of depressive disorders. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj19.515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Depression is a common mental disorder being one of the main causes of disability and mortality worldwide. Despite an intensive research during the past decades, the etiology of depressive disorders (DDs) remains incompletely understood; however, genetic factors are significantly involved in the liability to depression. The present review is focused on the studies based on a candidate gene approach, genome-wide association studies (GWAS) and whole exome sequencing (WES), which previously demonstrated associations between gene polymorphisms and DDs. According to the first approach, DD development is affected by serotonergic (TPH1, TPH2, HTR1A, HTR2A, and SLC6A4), dopaminergic (DRD4, SLC6A3) and noradrenergic (SLC6A2) system genes, and genes of enzymatic degradation (MAOA, COMT). In addition, there is evidence of the involvement of HPA-axis genes (OXTR, AVPR1A, and AVPR1B), sex hormone receptors genes (ESR1, ESR2, and AR), neurotrophin (BDNF) and methylenetetrahydrofolate reductase (MTHFR) genes, neuronal apoptosis (CASP3, BCL-XL, BAX, NPY, APP, and GRIN1) and inflammatory system (TNF, CRP, IL6, IL1B, PSMB4, PSMD9, and STAT3) genes in DD development. The results of the second approach (GWAS and WES) revealed that the PCLO, SIRT1, GNL3, GLT8D1, ITIH3, MTNR1A, BMP5, FHIT, KSR2, PCDH9, and AUTS2 genes predominantly responsible for neurogenesis and cell adhesion are involved in liability to depression. Therefore, the findings discussed suggest that genetic liability to DD is a complex process, which assumes simultaneous functioning of multiple genes including those reported previously, and requires future research in this field.
Collapse
Affiliation(s)
- Yu. D. Davydova
- Institute of Biochemistry and Genetics – Subdivision of the Ufa Federal Research Centre, RAS
| | - R. F. Enikeeva
- Institute of Biochemistry and Genetics – Subdivision of the Ufa Federal Research Centre, RAS
| | - A. V. Kazantseva
- Institute of Biochemistry and Genetics – Subdivision of the Ufa Federal Research Centre, RAS
| | - R. N. Mustafin
- Bashkir State University;
Bashkir State Medical University of the Ministry of Health of the Russian Federation
| | | | - S. B. Malykh
- Psychological Institute of Russian Academy of Education
| | - E. K. Khusnutdinov
- Institute of Biochemistry and Genetics – Subdivision of the Ufa Federal Research Centre, RAS;
Bashkir State University
| |
Collapse
|
20
|
TSPO upregulation in bipolar disorder and concomitant downregulation of mitophagic proteins and NLRP3 inflammasome activation. Neuropsychopharmacology 2019; 44:1291-1299. [PMID: 30575805 PMCID: PMC6785146 DOI: 10.1038/s41386-018-0293-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/24/2018] [Accepted: 12/03/2018] [Indexed: 01/04/2023]
Abstract
Bipolar disorder (BD) is a chronic, debilitating illness with a global prevalence of up to 4.8%. The importance of understanding how dysfunctional mitochondria and mitophagy contribute to cell survival and death in BD is becoming increasingly apparent. Therefore, the purpose of this study was to evaluate the mitophagic pathway and NLRP3 inflammasome activation in peripheral blood mononuclear cells (PBMCs) of patients with BD and healthy individuals. Since 18-kDa translocator protein (TSPO) plays an important role in regulating mitochondrial function and since TSPO itself impairs cellular mitophagy, we also investigated the changes in the TSPO-related pathway. Our results showed that patients with BD had lower levels of Parkin, p62/SQSTM1 and LC3A and an upregulation of TSPO pathway proteins (TSPO and VDAC), both in terms of mRNA and protein levels. Additionally, we found a negative correlation between mitophagy-related proteins and TSPO levels, while VDAC correlated negatively with p62/SQSTM1 and LC3 protein levels. Moreover, we found that the gene expression levels of the NLRP3-related proteins NLRP3, ASC, and pro-casp1 were upregulated in BD patients, followed by an increase in caspase-1 activity as well as IL-1β and IL-18 levels. As expected, there was a strong positive correlation between NLRP3-related inflammasome activation and TSPO-related proteins. The data reported here suggest that TSPO-VDAC complex upregulation in BD patients, the simultaneous downregulation of mitophagic proteins and NLRP3 inflammasome activation could lead to an accumulation of dysfunctional mitochondria, resulting in inflammation and apoptosis. In summary, the findings of this study provide novel evidence that mitochondrial dysfunction measured in peripheral blood is associated with BD.
Collapse
|
21
|
Albert PR, Le François B, Vahid-Ansari F. Genetic, epigenetic and posttranscriptional mechanisms for treatment of major depression: the 5-HT1A receptor gene as a paradigm. J Psychiatry Neurosci 2019; 44:164-176. [PMID: 30807072 PMCID: PMC6488484 DOI: 10.1503/jpn.180209] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/10/2018] [Accepted: 12/21/2018] [Indexed: 02/07/2023] Open
Abstract
Major depression and anxiety are highly prevalent and involve chronic dysregulation of serotonin, but they remain poorly understood. Here, we review novel transcriptional (genetic, epigenetic) and posttranscriptional (microRNA, alternative splicing) mechanisms implicated in mental illness, focusing on a key serotonin-related regulator, the serotonin 1A (5-HT1A) receptor. Functional single-nucleotide polymorphisms and stress-induced DNA methylation of the 5-HT1A promoter converge to differentially alter pre- and postsynaptic 5-HT1A receptor expression associated with major depression and reduced therapeutic response to serotonergic antidepressants. Major depression is also associated with altered levels of splice factors and microRNA, posttranscriptional mechanisms that regulate RNA stability. The human 5-HT1A 3′-untranslated region is alternatively spliced, removing microRNA sites and increasing 5-HT1A expression, which is reduced in major depression and may be genotype-dependent. Thus, the 5-HT1A receptor gene illustrates the convergence of genetic, epigenetic and posttranscriptional mechanisms in gene expression, neurodevelopment and neuroplasticity, and major depression. Understanding gene regulatory mechanisms could enhance the detection, categorization and personalized treatment of major depression.
Collapse
Affiliation(s)
- Paul R. Albert
- From the Department of Neuroscience, Ottawa Hospital Research Institute, UOttawa Brain and Mind Research Institute, Ottawa, Ont., Canada
| | - Brice Le François
- From the Department of Neuroscience, Ottawa Hospital Research Institute, UOttawa Brain and Mind Research Institute, Ottawa, Ont., Canada
| | - Faranak Vahid-Ansari
- From the Department of Neuroscience, Ottawa Hospital Research Institute, UOttawa Brain and Mind Research Institute, Ottawa, Ont., Canada
| |
Collapse
|
22
|
Kahl KG, Stapel B, Frieling H. Link between depression and cardiovascular diseases due to epigenomics and proteomics: Focus on energy metabolism. Prog Neuropsychopharmacol Biol Psychiatry 2019; 89:146-157. [PMID: 30194950 DOI: 10.1016/j.pnpbp.2018.09.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/13/2018] [Accepted: 09/05/2018] [Indexed: 12/11/2022]
Abstract
Major depression is the most common mental disorder and a leading cause of years lived with disability. In addition to the burden attributed to depressive symptoms and reduced daily life functioning, people with major depression are at increased risk of premature mortality, particularly due to cardiovascular diseases. Several studies point to a bi-directional relation between major depression and cardiovascular diseases, thereby indicating that both diseases may share common pathophysiological pathways. These include lifestyle factors (e.g. physical activity, smoking behavior), dysfunctions of endocrine systems (e.g. hypothalamus-pituitary adrenal axis), and a dysbalance of pro- and anti-inflammatory factors. Furthermore, recent research point to the role of epigenomic and proteomic factors, that are reviewed here with a particular focus on the mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Kai G Kahl
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Germany.
| | - Britta Stapel
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Germany
| | - Helge Frieling
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Germany
| |
Collapse
|
23
|
Kautzky A, James GM, Philippe C, Baldinger-Melich P, Kraus C, Kranz GS, Vanicek T, Gryglewski G, Hartmann AM, Hahn A, Wadsak W, Mitterhauser M, Rujescu D, Kasper S, Lanzenberger R. Epistasis of HTR1A and BDNF risk genes alters cortical 5-HT1A receptor binding: PET results link genotype to molecular phenotype in depression. Transl Psychiatry 2019; 9:5. [PMID: 30664620 PMCID: PMC6341100 DOI: 10.1038/s41398-018-0308-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 07/31/2018] [Indexed: 02/07/2023] Open
Abstract
Alterations of the 5-HT1A receptor and BDNF have consistently been associated with affective disorders. Two functional single nucleotide polymorphisms (SNPs), rs6295 of the serotonin 1A receptor gene (HTR1A) and rs6265 of brain-derived neurotrophic factor gene (BDNF), may impact transcriptional regulation and expression of the 5-HT1A receptor. Here we investigated interaction effects of rs6295 and rs6265 on 5-HT1A receptor binding. Forty-six healthy subjects were scanned with PET using the radioligand [carbonyl-11C]WAY-100635. Genotyping was performed for rs6265 and rs6295. Subjects showing a genotype with at least three risk alleles (G of rs6295 or A of rs6265) were compared to control genotypes. Cortical surface binding potential (BPND) was computed for 32 cortical regions of interest (ROI). Mixed model was applied to study main and interaction effects of ROI and genotype. ANOVA was used for post hoc analyses. Individuals with the risk genotypes exhibited an increase in 5-HT1A receptor binding by an average of 17% (mean BPND 3.56 ± 0.74 vs. 2.96 ± 0.88). Mixed model produced an interaction effect of ROI and genotype on BPND and differences could be demonstrated in 10 ROI post hoc. The combination of disadvantageous allelic expression of rs6295 and rs6265 may result in a 5-HT1A receptor profile comparable to affective disorders as increased 5-HT1A receptor binding is a well published phenotype of depression. Thus, epistasis between BDNF and HTR1A may contribute to the multifactorial risk for affective disorders and our results strongly advocate further research on this genetic signature in affective disorders.
Collapse
Affiliation(s)
- Alexander Kautzky
- 0000 0000 9259 8492grid.22937.3dDepartment of Psychiatry and Psychotherapy, Medical University of Vienna, Wien, Austria
| | - Gregory M. James
- 0000 0000 9259 8492grid.22937.3dDepartment of Psychiatry and Psychotherapy, Medical University of Vienna, Wien, Austria
| | - Cecile Philippe
- 0000 0000 9259 8492grid.22937.3dDivision of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Wien, Austria
| | - Pia Baldinger-Melich
- 0000 0000 9259 8492grid.22937.3dDepartment of Psychiatry and Psychotherapy, Medical University of Vienna, Wien, Austria
| | - Christoph Kraus
- 0000 0000 9259 8492grid.22937.3dDepartment of Psychiatry and Psychotherapy, Medical University of Vienna, Wien, Austria
| | - Georg S. Kranz
- 0000 0000 9259 8492grid.22937.3dDepartment of Psychiatry and Psychotherapy, Medical University of Vienna, Wien, Austria
| | - Thomas Vanicek
- 0000 0000 9259 8492grid.22937.3dDepartment of Psychiatry and Psychotherapy, Medical University of Vienna, Wien, Austria
| | - Gregor Gryglewski
- 0000 0000 9259 8492grid.22937.3dDepartment of Psychiatry and Psychotherapy, Medical University of Vienna, Wien, Austria
| | - Annette M. Hartmann
- 0000 0001 0679 2801grid.9018.0University Clinic for Psychiatry, Psychotherapy and Psychosomatic, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Andreas Hahn
- 0000 0000 9259 8492grid.22937.3dDepartment of Psychiatry and Psychotherapy, Medical University of Vienna, Wien, Austria
| | - Wolfgang Wadsak
- 0000 0000 9259 8492grid.22937.3dDivision of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Wien, Austria ,grid.499898.dCenter for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Markus Mitterhauser
- 0000 0000 9259 8492grid.22937.3dDivision of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Wien, Austria ,Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Dan Rujescu
- 0000 0001 0679 2801grid.9018.0University Clinic for Psychiatry, Psychotherapy and Psychosomatic, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Siegfried Kasper
- 0000 0000 9259 8492grid.22937.3dDepartment of Psychiatry and Psychotherapy, Medical University of Vienna, Wien, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Wien, Austria.
| |
Collapse
|
24
|
Wang H, Yin F, Gao J, Fan X. Association Between 5-HTTLPR Polymorphism and the Risk of Autism: A Meta-Analysis Based on Case-Control Studies. Front Psychiatry 2019; 10:51. [PMID: 30814960 PMCID: PMC6381045 DOI: 10.3389/fpsyt.2019.00051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/24/2019] [Indexed: 11/13/2022] Open
Abstract
Background: Recently, many case-control studies have reported the association between 5-HTTLPR polymorphism and autism risk. However, the results are inconclusive and conflicting. To investigate the genetic association of 5-HTTLPR polymorphism and autism risk, we conducted a comprehensive meta-analysis based on previous case-control studies. Methods: Literature search was performed through PubMed, Embase, Web of Knowledge and CNKI databases until June 27, 2018. The strength of the association was assessed by relative risk (RR) and its corresponding 95% confidence interval (CI). Fixed or random effect model was selected based on the results of heterogeneity test. Further, subgroup analyses were conducted to explore the association of 5-HTTLPR polymorphism and autism risk in different population. Results: Eleven studies with 930 cases and 1234 controls were identified. Although there was a significant association between 5-HTTLPR polymorphism and autism risk under the dominant model after removing the studies causing heterogeneity, the significance did not exist after Bonferroni's correction. Subgroup analyses also showed similar results after Bonferroni's correction. In addition, there was no obvious publication bias in our meta-analysis. Conclusions: Our present meta-analysis does not support a direct effect of 5-HTTLPR polymorphism on autism risk according to present results. Further analyses of the effect of genetic networks and more well designed studies with larger sample size are required.
Collapse
Affiliation(s)
- Hongbing Wang
- Department of Radiotherapy Oncology, Cangzhou Central Hospital, Cangzhou, China
| | - Fangna Yin
- Clinical Laboratory, Cangzhou Central Hospital, Cangzhou, China
| | - Junwei Gao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| |
Collapse
|
25
|
Abstract
Stress is an adaptive response to environment aversive stimuli and a common life experience of one's daily life. Chronic or excessive stress especially that happened in early life is found to be deleterious to individual's physical and mental health, which is highly related to depressive disorders onset. Stressful life events are consistently considered to be the high-risk factors of environment for predisposing depressive disorders. In linking stressful life events with depressive disorder onset, dysregulated HPA axis activity is supposed to play an important role in mediating aversive impacts of life stress on brain structure and function. Increasing evidence have indicated the strong association of stress, especially the chronic stress and early life stress, with depressive disorders development, while the association of stress with depression is moderated by genetic risk factors, including polymorphism of SERT, BDNF, GR, FKBP5, MR, and CRHR1. Meanwhile, stressful life experience particularly early life stress will exert epigenetic modification in these risk genes via DNA methylation and miRNA regulation to generate long-lasting effects on these genes expression, which in turn cause brain structural and functional alteration, and finally increase the vulnerability to depressive disorders. Therefore, the interaction of environment with gene, in which stressful life exposure interplay with genetic risk factors and epigenetic modification, is essential in predicting depressive disorders development. As the mediator of environmental risk factors, stress will function together with genetic and epigenetic mechanism to influence brain structure and function, physiology and psychology, and finally the vulnerability to depressive disorders.
Collapse
|
26
|
A Novel Alternative Splicing Mechanism That Enhances Human 5-HT1A Receptor RNA Stability Is Altered in Major Depression. J Neurosci 2018; 38:8200-8210. [PMID: 30093565 DOI: 10.1523/jneurosci.0902-18.2018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/10/2018] [Accepted: 07/17/2018] [Indexed: 11/21/2022] Open
Abstract
The serotonin-1A (5-HT1A) receptor is a key regulator of serotonergic activity and is implicated in mood and emotion. However, its post-transcriptional regulation has never been studied in humans. In the present study, we show that the "intronless" human 5-HT1A gene (HTR1A) is alternatively spliced in its 3'-UTR, yielding two novel splice variants. These variants lack a ∼1.6 kb intron, which contains an microRNA-135 (miR135) target site. Unlike the human HTR1A, the mouse HTR1A lacks the splice donor/accepter sites. Thus, in the mouse HTR1A, splicing was not detected. The two spliced mRNAs are extremely stable, are resistant to miR135-induced downregulation, and have greater translational output than the unspliced variant. Moreover, alternative HTR1A RNA splicing is oppositely regulated by the splice factors PTBP1 and nSR100, which inhibit or enhance its splicing, respectively. In postmortem human brain tissue from both sexes, HTR1A mRNA splicing was prevalent and region-specific. Unspliced HTR1A was expressed more strongly in the hippocampus and midbrain versus the prefrontal cortex (PFC), and correlated with reduced levels of nSR100. Importantly, HTR1A RNA splicing and nSR100 levels were reduced in the PFC of individuals with major depression compared with controls. Our unexpected findings uncover a novel mechanism to regulate HTR1A gene expression through alternative splicing of microRNA sites. Altered levels of splice factors could contribute to changes in regional and depression-related gene expression through alternative splicing.SIGNIFICANCE STATEMENT Alternative splicing, which is prevalent in brain tissue, increases gene diversity. The serotonin-1A receptor gene (HTR1A) is a regulator of serotonin, which is implicated in mood and emotion. Here we show that human HTR1A RNA is alternately spliced. Splicing removes a microRNA site to generate ultrastable RNA and increase HTR1A expression. This splicing varies in different brain regions and is reduced in major depression. We also identify specific splice factors for HTR1A RNA, showing they are also reduced in depression. Thus, we describe a novel mechanism to regulate gene expression through splicing. Altered levels of splice factors could contribute to depression by changing gene expression.
Collapse
|
27
|
Philippe TJ, Vahid-Ansari F, Donaldson ZR, Le François B, Zahrai A, Turcotte-Cardin V, Daigle M, James J, Hen R, Merali Z, Albert PR. Loss of MeCP2 in adult 5-HT neurons induces 5-HT1A autoreceptors, with opposite sex-dependent anxiety and depression phenotypes. Sci Rep 2018; 8:5788. [PMID: 29636529 PMCID: PMC5893553 DOI: 10.1038/s41598-018-24167-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/27/2018] [Indexed: 12/11/2022] Open
Abstract
The 5-HT1A autoreceptor mediates feedback inhibition of serotonin (5-HT) neurons, and is implicated in major depression. The human 5-HT1A gene (HTR1A) rs6295 risk allele prevents Deaf1 binding to HTR1A, resulting in increased 5-HT1A autoreceptor transcription. Since chronic stress alters HTR1A methylation and expression, we addressed whether recruitment of methyl-binding protein MeCP2 may alter Deaf1 regulation at the HTR1A locus. We show that MeCP2 enhances Deaf1 binding to its HTR1A site and co-immunoprecipitates with Deaf1 in cells and brain tissue. Chromatin immunoprecipitation assays showed Deaf1-dependent recruitment of MeCP2 to the mouse HTR1A promoter, and MeCP2 modulated human and mouse HTR1A gene transcription in a Deaf1-dependent fashion, enhancing Deaf1-induced repression at the Deaf1 site. To address the role of MeCP2 in HTR1A regulation in vivo, mice with conditional knockout of MeCP2 in adult 5-HT neurons (MeCP2 cKO) were generated. These mice exhibited increased 5-HT1A autoreceptor levels and function, consistent with MeCP2 enhancement of Deaf1 repression in 5-HT neurons. Interestingly, female MeCP2-cKO mice displayed reduced anxiety, while males showed increased anxiety and reduced depression-like behaviors. These data uncover a novel role for MeCP2 in 5-HT neurons to repress HTR1A expression and drive adult anxiety- and depression-like behaviors in a sex-specific manner.
Collapse
Affiliation(s)
- Tristan J Philippe
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Faranak Vahid-Ansari
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Zoe R Donaldson
- Department of Molecular, Cellular, and Developmental Biology and Department of Psychology & Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Brice Le François
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Amin Zahrai
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Valérie Turcotte-Cardin
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Mireille Daigle
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Jonathan James
- The Royal's Institute of Mental Health, affiliated with the University of Ottawa, Ottawa, ON, Canada
| | - René Hen
- New York State Psychiatric Institute, Columbia University Medical Center and Research Foundation for Mental Hygiene, New York, NY, USA.,Department of Psychiatry, Columbia University, New York, NY, USA
| | - Zul Merali
- The Royal's Institute of Mental Health, affiliated with the University of Ottawa, Ottawa, ON, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada. .,Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
28
|
Diamantopoulou A, Kalpachidou T, Aspiotis G, Gampierakis I, Stylianopoulou F, Stamatakis A. An early experience of mild adversity involving temporary denial of maternal contact affects the serotonergic system of adult male rats and leads to a depressive-like phenotype and inability to adapt to a chronic social stress. Physiol Behav 2018; 184:46-54. [DOI: 10.1016/j.physbeh.2017.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/09/2017] [Accepted: 11/06/2017] [Indexed: 10/18/2022]
|
29
|
Vai B, Riberto M, Ghiglino D, Poletti S, Bollettini I, Lorenzi C, Colombo C, Benedetti F. A 5-HT 1Areceptor promoter polymorphism influences fronto-limbic functional connectivity and depression severity in bipolar disorder. Psychiatry Res Neuroimaging 2017; 270:1-7. [PMID: 28985530 DOI: 10.1016/j.pscychresns.2017.09.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 09/18/2017] [Accepted: 09/18/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Benedetta Vai
- IRCCS Ospedale San Raffaele, Department of Clinical Neurosciences, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), University Vita-Salute San Raffaele, Milan, Italy.
| | - Martina Riberto
- IRCCS Ospedale San Raffaele, Department of Clinical Neurosciences, Milan, Italy
| | - Davide Ghiglino
- IRCCS Ospedale San Raffaele, Department of Clinical Neurosciences, Milan, Italy
| | - Sara Poletti
- IRCCS Ospedale San Raffaele, Department of Clinical Neurosciences, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), University Vita-Salute San Raffaele, Milan, Italy
| | - Irene Bollettini
- IRCCS Ospedale San Raffaele, Department of Clinical Neurosciences, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), University Vita-Salute San Raffaele, Milan, Italy
| | - Cristina Lorenzi
- IRCCS Ospedale San Raffaele, Department of Clinical Neurosciences, Milan, Italy
| | - Cristina Colombo
- IRCCS Ospedale San Raffaele, Department of Clinical Neurosciences, Milan, Italy
| | - Francesco Benedetti
- IRCCS Ospedale San Raffaele, Department of Clinical Neurosciences, Milan, Italy; C.E.R.M.A.C. (Centro di Eccellenza Risonanza Magnetica ad Alto Campo), University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
30
|
Cai CY, Wu HY, Luo CX, Zhu DY, Zhang Y, Zhou QG, Zhang J. Extracellular regulated protein kinaseis critical for the role of 5-HT1a receptor in modulating nNOS expression and anxiety-related behaviors. Behav Brain Res 2017; 357-358:88-97. [PMID: 29246772 DOI: 10.1016/j.bbr.2017.12.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/04/2017] [Accepted: 12/11/2017] [Indexed: 12/20/2022]
Abstract
Our previous study found that serotonin 1A receptor (5-HT1aR) is an endogenous suppressor of nNOS expression in the hippocampus, which accounts for anxiolytic effect of fluoxetine. However, the precise molecular mechanism remains unknown. By using 8-OH-DPAT, a selective 5-HT1aR agonist, NAN-190, a selective 5-HT1aR antagonist, and U0126, an Extracellular Regulated Protein Kinases (ERK) phosphorylation inhibitor, we investigated the role of ERK in 5-HT1aR-nNOS pathway. Western blots analysis demonstrated that 5-HT1aR activation up-regulated the level of phosphorylated ERK (P-ERK) beginning at 5 min and down-regulated the expression of nNOS beginning at 20 min. Meanwhile, blockage of 5-HT1aR resulted in a decrease in P-ERK beginning at 20 min and caused an increase in nNOS expression beginning at 6 h. Although U0126 itself did not alter nNOS expression and activity, NO level, and anxiety-related behaviors, the treatment totally reversed 8-OH-DPAT-induced reduction in nNOS expression and function, and anxiolytic effect. Besides, our data showed that ERK phosphorylation was essential for 5-HT1aR activation-induced cAMP responsive element binding protein (CREB) phosphorylation, hippocampal neurogenesis and synaptogenesis of newborn neuron. Our study suggests a crucial role of ERK phosphorylation in the regulation of nNOS expression by 5-HT1aR, which is helpful for understanding the mechanism of 5-HT1aR-based anxiolytic treatment.
Collapse
Affiliation(s)
- Cheng-Yun Cai
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Hai-Yin Wu
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Chun-Xia Luo
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Dong-Ya Zhu
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China; The Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing, 211166, People's Republic of China
| | - Yu Zhang
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China.
| | - Qi-Gang Zhou
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China.
| | - Jing Zhang
- Institution of Stem Cells and Neuroregeneration, Nanjing Medical University, Nanjing, 211166, People's Republic of China; Departments of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, People's Republic of China.
| |
Collapse
|
31
|
Abrogated Freud-1/Cc2d1a Repression of 5-HT1A Autoreceptors Induces Fluoxetine-Resistant Anxiety/Depression-Like Behavior. J Neurosci 2017; 37:11967-11978. [PMID: 29101244 DOI: 10.1523/jneurosci.1668-17.2017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 09/29/2017] [Accepted: 10/10/2017] [Indexed: 11/21/2022] Open
Abstract
Freud-1/Cc2d1a represses the gene transcription of serotonin-1A (5-HT1A) autoreceptors, which negatively regulate 5-HT tone. To test the role of Freud-1 in vivo, we generated mice with adulthood conditional knock-out of Freud-1 in 5-HT neurons (cF1ko). In cF1ko mice, 5-HT1A autoreceptor protein, binding and hypothermia response were increased, with reduced 5-HT content and neuronal activity in the dorsal raphe. The cF1ko mice displayed increased anxiety- and depression-like behavior that was resistant to chronic antidepressant (fluoxetine) treatment. Using conditional Freud-1/5-HT1A double knock-out (cF1/1A dko) to disrupt both Freud-1 and 5-HT1A genes in 5-HT neurons, no increase in anxiety- or depression-like behavior was seen upon knock-out of Freud-1 on the 5-HT1A autoreceptor-negative background; rather, a reduction in depression-like behavior emerged. These studies implicate transcriptional dysregulation of 5-HT1A autoreceptors by the repressor Freud-1 in anxiety and depression and provide a clinically relevant genetic model of antidepressant resistance. Targeting specific transcription factors, such as Freud-1, to restore transcriptional balance may augment response to antidepressant treatment.SIGNIFICANCE STATEMENT Altered regulation of the 5-HT1A autoreceptor has been implicated in human anxiety, major depression, suicide, and resistance to antidepressants. This study uniquely identifies a single transcription factor, Freud-1, as crucial for 5-HT1A autoreceptor expression in vivo Disruption of Freud-1 in serotonin neurons in mice links upregulation of 5-HT1A autoreceptors to anxiety/depression-like behavior and provides a new model of antidepressant resistance. Treatment strategies to reestablish transcriptional regulation of 5-HT1A autoreceptors could provide a more robust and sustained antidepressant response.
Collapse
|
32
|
Vadodaria KC, Stern S, Marchetto MC, Gage FH. Serotonin in psychiatry: in vitro disease modeling using patient-derived neurons. Cell Tissue Res 2017; 371:161-170. [PMID: 28812143 DOI: 10.1007/s00441-017-2670-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 07/01/2017] [Indexed: 01/18/2023]
Abstract
Several lines of evidence implicate serotonin in the etiology of multiple psychiatric disorders, especially mood disorders, such as major depressive disorder (MDD) and bipolar disorder (BD). Much of our current understanding of biological mechanisms underlying serotonergic alterations in mood disorders comes from animal studies. Innovation in induced pluripotent stem cell and transdifferentiation technologies for deriving neurons from adult humans has enabled the study of disease-relevant cellular phenotypes in vitro. In this context, human serotonergic neurons can now be generated using three recently published methodologies. In this mini-review, we broadly discuss evidence linking altered serotonergic neurotransmission in MDD and BD and focus on recently published methods for generating human serotonergic neurons in vitro.
Collapse
Affiliation(s)
- Krishna C Vadodaria
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, USA
| | - Shani Stern
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, USA
| | - Maria C Marchetto
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, USA
| | - Fred H Gage
- Laboratory of Genetics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, USA.
| |
Collapse
|
33
|
Abstract
Depression is a polygenic and highly complex psychiatric disorder that remains a major burden on society. Antidepressants, such as selective serotonin reuptake inhibitors (SSRIs), are some of the most commonly prescribed drugs worldwide. In this review, we will discuss the evidence that links serotonin and serotonin receptors to the etiology of depression and the mechanisms underlying response to antidepressant treatment. We will then revisit the role of serotonin in three distinct hypotheses that have been proposed over the last several decades to explain the pathophysiology of depression: the monoamine, neurotrophic, and neurogenic hypotheses. Finally, we will discuss how recent studies into serotonin receptors have implicated specific neural circuitry in mediating the antidepressant response, with a focus being placed on the hippocampus.
Collapse
Affiliation(s)
- Christine N Yohn
- Department of Psychology, Behavioral & Systems Neuroscience Area, Rutgers, The State University of New Jersey, 152 Frelinghuysen Rd., Room 215, Piscataway, NJ, 08816, USA
| | - Mark M Gergues
- Department of Psychology, Behavioral & Systems Neuroscience Area, Rutgers, The State University of New Jersey, 152 Frelinghuysen Rd., Room 215, Piscataway, NJ, 08816, USA
| | - Benjamin Adam Samuels
- Department of Psychology, Behavioral & Systems Neuroscience Area, Rutgers, The State University of New Jersey, 152 Frelinghuysen Rd., Room 215, Piscataway, NJ, 08816, USA.
| |
Collapse
|
34
|
Higgins GA, Allyn-Feuer A, Georgoff P, Nikolian V, Alam HB, Athey BD. Mining the topography and dynamics of the 4D Nucleome to identify novel CNS drug pathways. Methods 2017; 123:102-118. [PMID: 28385536 DOI: 10.1016/j.ymeth.2017.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/10/2017] [Indexed: 12/16/2022] Open
Abstract
The pharmacoepigenome can be defined as the active, noncoding province of the genome including canonical spatial and temporal regulatory mechanisms of gene regulation that respond to xenobiotic stimuli. Many psychotropic drugs that have been in clinical use for decades have ill-defined mechanisms of action that are beginning to be resolved as we understand the transcriptional hierarchy and dynamics of the nucleus. In this review, we describe spatial, temporal and biomechanical mechanisms mediated by psychotropic medications. Focus is placed on a bioinformatics pipeline that can be used both for detection of pharmacoepigenomic variants that discretize drug response and adverse events to improve pharmacogenomic testing, and for the discovery of novel CNS therapeutics. This approach integrates the functional topology and dynamics of the transcriptional hierarchy of the pharmacoepigenome, gene variant-driven identification of pharmacogenomic regulatory domains, and mesoscale mapping for the discovery of novel CNS pharmacodynamic pathways in human brain. Examples of the application of this pipeline are provided, including the discovery of valproic acid (VPA) mediated transcriptional reprogramming of neuronal cell fate following injury, and mapping of a CNS pathway glutamatergic pathway for the mood stabilizer lithium. These examples in regulatory pharmacoepigenomics illustrate how ongoing research using the 4D nucleome provides a foundation to further insight into previously unrecognized psychotropic drug pharmacodynamic pathways in the human CNS.
Collapse
Affiliation(s)
- Gerald A Higgins
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, USA
| | - Ari Allyn-Feuer
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, USA
| | - Patrick Georgoff
- Department of Surgery, University of Michigan Medical School, USA
| | - Vahagn Nikolian
- Department of Surgery, University of Michigan Medical School, USA
| | - Hasan B Alam
- Department of Surgery, University of Michigan Medical School, USA
| | - Brian D Athey
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, USA; Michigan Institute for Data Science (MIDAS), USA.
| |
Collapse
|
35
|
Anderson MR, Miller L, Wickramaratne P, Svob C, Odgerel Z, Zhao R, Weissman MM. Genetic Correlates of Spirituality/Religion and Depression: A Study in Offspring and Grandchildren at High and Low Familial Risk for Depression. ACTA ACUST UNITED AC 2017; 4:43-63. [PMID: 29057276 DOI: 10.1037/scp0000125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
RATIONALE Possible genetic correlates of spirituality and depression have been identified in community samples. We investigate some of the previously identified candidates in a sample of families at both high and low-risk for depression. METHOD Offspring and grandchildren of individuals at high and low-risk for depression, participating in a multi-wave thirty-year longitudinal study, were assessed for seven SNPS drawn from four single gene candidates associated with systems implicated in both depression and spirituality: Serotonin (5-HT1B and 5-HT2A), Dopamine (DRD2), Oxytocin (OT) and Monoamine Vesicular Transporter (VMAT1). RESULTS Dopamine (DRD2) Serotonin (5-HT1B), their Transporter (VMAT1) and Oxytocin (OXTR) were positively associated with a high level of importance of spirituality or religion (S/R) in the group at low familial risk for depression. DRD2 minor allele was associated with both lifetime major depressive disorder (MDD) and spirituality in the low-risk group for depression. No SNPs were related to S/R in the group at high familial risk for depression. OXTR was associated with lifetime MDD in the full sample. CONCLUSION Genes for dopamine, serotonin, their vesicular transporter, and oxytocin may be associated with S/R in people at low familial risk for depression. Genes for dopamine may be associated both with S/R and increased risk for depression in people at low-risk for depression, suggesting a common pathway or physiology to mild to moderate depression. MDD is associated with oxytocin across risk groups. In the high-risk group, phenotypic expression of S/R may be suppressed. IMPLICATIONS The shared association of DRD2 by S/R and depression, generally found to be inversely related, calls for further research on their common physiological pathways, and the phenotypic expression of these pathways based upon use and environment. Prevention for offspring at high familial risk for depression might include support for the development of child spirituality.
Collapse
Affiliation(s)
| | - Lisa Miller
- Teachers College, Columbia University, New York, NY
| | - Priya Wickramaratne
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Division of Epidemiology, New York State Psychiatric Institute, New York, NY, USA.,Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Connie Svob
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Division of Epidemiology, New York State Psychiatric Institute, New York, NY, USA
| | - Zagaa Odgerel
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Division of Epidemiology, New York State Psychiatric Institute, New York, NY, USA
| | - Ruixin Zhao
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Division of Epidemiology, New York State Psychiatric Institute, New York, NY, USA
| | - Myrna M Weissman
- Department of Psychiatry, College of Physicians and Surgeons, Columbia University, New York, NY, USA.,Division of Epidemiology, New York State Psychiatric Institute, New York, NY, USA.,Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
36
|
Fabbri C, Hosak L, Mössner R, Giegling I, Mandelli L, Bellivier F, Claes S, Collier DA, Corrales A, Delisi LE, Gallo C, Gill M, Kennedy JL, Leboyer M, Lisoway A, Maier W, Marquez M, Massat I, Mors O, Muglia P, Nöthen MM, O'Donovan MC, Ospina-Duque J, Propping P, Shi Y, St Clair D, Thibaut F, Cichon S, Mendlewicz J, Rujescu D, Serretti A. Consensus paper of the WFSBP Task Force on Genetics: Genetics, epigenetics and gene expression markers of major depressive disorder and antidepressant response. World J Biol Psychiatry 2017; 18:5-28. [PMID: 27603714 DOI: 10.1080/15622975.2016.1208843] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 06/29/2016] [Indexed: 12/11/2022]
Abstract
Major depressive disorder (MDD) is a heritable disease with a heavy personal and socio-economic burden. Antidepressants of different classes are prescribed to treat MDD, but reliable and reproducible markers of efficacy are not available for clinical use. Further complicating treatment, the diagnosis of MDD is not guided by objective criteria, resulting in the risk of under- or overtreatment. A number of markers of MDD and antidepressant response have been investigated at the genetic, epigenetic, gene expression and protein levels. Polymorphisms in genes involved in antidepressant metabolism (cytochrome P450 isoenzymes), antidepressant transport (ABCB1), glucocorticoid signalling (FKBP5) and serotonin neurotransmission (SLC6A4 and HTR2A) were among those included in the first pharmacogenetic assays that have been tested for clinical applicability. The results of these investigations were encouraging when examining patient-outcome improvement. Furthermore, a nine-serum biomarker panel (including BDNF, cortisol and soluble TNF-α receptor type II) showed good sensitivity and specificity in differentiating between MDD and healthy controls. These first diagnostic and response-predictive tests for MDD provided a source of optimism for future clinical applications. However, such findings should be considered very carefully because their benefit/cost ratio and clinical indications were not clearly demonstrated. Future tests may include combinations of different types of biomarkers and be specific for MDD subtypes or pathological dimensions.
Collapse
Affiliation(s)
- Chiara Fabbri
- a Department of Biomedical and Neuromotor Sciences , University of Bologna , Bologna , Italy
| | - Ladislav Hosak
- b Department of Psychiatrics , Charles University, Faculty of Medicine and University Hospital, Hradec Králové , Czech Republic
| | - Rainald Mössner
- c Department of Psychiatry and Psychotherapy , University of Tübingen , Tübingen , Germany
| | - Ina Giegling
- d Department of Psychiatry, Psychotherapy and Psychosomatics , Martin Luther University of Halle-Wittenberg , Halle , Germany
| | - Laura Mandelli
- a Department of Biomedical and Neuromotor Sciences , University of Bologna , Bologna , Italy
| | - Frank Bellivier
- e Fondation Fondamental, Créteil, France AP-HP , GH Saint-Louis-Lariboisière-Fernand-Widal, Pôle Neurosciences , Paris , France
| | - Stephan Claes
- f GRASP-Research Group, Department of Neuroscience , University of Leuven , Leuven , Belgium
| | - David A Collier
- g Social, Genetic and Developmental Psychiatry Centre , Institute of Psychiatry, King's College London , London , UK
| | - Alejo Corrales
- h National University (UNT) Argentina, Argentinean Association of Biological Psychiatry , Buenos Aires , Argentina
| | - Lynn E Delisi
- i VA Boston Health Care System , Brockton , MA , USA
| | - Carla Gallo
- j Departamento de Ciencias Celulares y Moleculares, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía , Universidad Peruana Cayetano Heredia , Lima , Peru
| | - Michael Gill
- k Neuropsychiatric Genetics Research Group, Department of Psychiatry , Trinity College Dublin , Dublin , Ireland
| | - James L Kennedy
- l Neurogenetics Section, Centre for Addiction and Mental Health , Toronto , Ontario , Canada
| | - Marion Leboyer
- m Faculté de Médecine , Université Paris-Est Créteil, Inserm U955, Equipe Psychiatrie Translationnelle , Créteil , France
| | - Amanda Lisoway
- l Neurogenetics Section, Centre for Addiction and Mental Health , Toronto , Ontario , Canada
| | - Wolfgang Maier
- n Department of Psychiatry , University of Bonn , Bonn , Germany
| | - Miguel Marquez
- o Director of ADINEU (Asistencia, Docencia e Investigación en Neurociencia) , Buenos Aires , Argentina
| | - Isabelle Massat
- p UNI - ULB Neurosciences Institute, ULB , Bruxelles , Belgium
| | - Ole Mors
- q Department P , Aarhus University Hospital , Risskov , Denmark
| | | | - Markus M Nöthen
- s Institute of Human Genetics , University of Bonn , Bonn , Germany
| | - Michael C O'Donovan
- t MRC Centre for Neuropsychiatric Genetics and Genomics , Institute of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University , Cardiff , UK
| | - Jorge Ospina-Duque
- u Grupo de Investigación en Psiquiatría, Departamento de Psiquiatría, Facultad de Medicina , Universidad de Antioquia , Medellín , Colombia
| | | | - Yongyong Shi
- w Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education , Shanghai Jiao Tong University , Shanghai , China
| | - David St Clair
- x University of Aberdeen, Institute of Medical Sciences , Aberdeen , UK
| | - Florence Thibaut
- y University Hospital Cochin (Site Tarnier), University Sorbonne Paris Cité (Faculty of Medicine Paris Descartes), INSERM U 894 Centre Psychiatry and Neurosciences , Paris , France
| | - Sven Cichon
- z Division of Medical Genetics, Department of Biomedicine , University of Basel , Basel , Switzerland
| | - Julien Mendlewicz
- aa Laboratoire de Psychologie Medicale, Centre Européen de Psychologie Medicale , Université Libre de Bruxelles and Psy Pluriel , Brussels , Belgium
| | - Dan Rujescu
- d Department of Psychiatry, Psychotherapy and Psychosomatics , Martin Luther University of Halle-Wittenberg , Halle , Germany
| | - Alessandro Serretti
- a Department of Biomedical and Neuromotor Sciences , University of Bologna , Bologna , Italy
| |
Collapse
|
37
|
Davis MT, Holmes SE, Pietrzak RH, Esterlis I. Neurobiology of Chronic Stress-Related Psychiatric Disorders: Evidence from Molecular Imaging Studies. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2017; 1:2470547017710916. [PMID: 29862379 PMCID: PMC5976254 DOI: 10.1177/2470547017710916] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/30/2017] [Accepted: 05/01/2017] [Indexed: 01/12/2023]
Abstract
Chronic stress accounts for billions of dollars of economic loss annually in the United States alone, and is recognized as a major source of disability and mortality worldwide. Robust evidence suggests that chronic stress plays a significant role in the onset of severe and impairing psychiatric conditions, including major depressive disorder, bipolar disorder, and posttraumatic stress disorder. Application of molecular imaging techniques such as positron emission tomography and single photon emission computed tomography in recent years has begun to provide insight into the molecular mechanisms by which chronic stress confers risk for these disorders. The present paper provides a comprehensive review and synthesis of all positron emission tomography and single photon emission computed tomography imaging publications focused on the examination of molecular targets in individuals with major depressive disorder, posttraumatic stress disorder, or bipolar disorder to date. Critical discussion of discrepant findings and broad strengths and weaknesses of the current body of literature is provided. Recommended future directions for the field of molecular imaging to further elucidate the neurobiological substrates of chronic stress-related disorders are also discussed. This article is part of the inaugural issue for the journal focused on various aspects of chronic stress.
Collapse
Affiliation(s)
- Margaret T. Davis
- Department of Psychiatry, Yale School of
Medicine, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical
Imaging, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Sophie E. Holmes
- Department of Psychiatry, Yale School of
Medicine, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical
Imaging, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Robert H. Pietrzak
- Department of Psychiatry, Yale School of
Medicine, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical
Imaging, Yale School of Medicine, Yale University, New Haven, CT, USA
- US Department of Veterans Affairs National
Center for Posttraumatic Stress Disorder, VA Connecticut Healthcare System, West Haven, CT,
USA
| | - Irina Esterlis
- Department of Psychiatry, Yale School of
Medicine, Yale University, New Haven, CT, USA
- Department of Radiology and Biomedical
Imaging, Yale School of Medicine, Yale University, New Haven, CT, USA
- US Department of Veterans Affairs National
Center for Posttraumatic Stress Disorder, VA Connecticut Healthcare System, West Haven, CT,
USA
| |
Collapse
|
38
|
Amare AT, Schubert KO, Klingler-Hoffmann M, Cohen-Woods S, Baune BT. The genetic overlap between mood disorders and cardiometabolic diseases: a systematic review of genome wide and candidate gene studies. Transl Psychiatry 2017; 7:e1007. [PMID: 28117839 PMCID: PMC5545727 DOI: 10.1038/tp.2016.261] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 10/21/2016] [Accepted: 10/31/2016] [Indexed: 12/11/2022] Open
Abstract
Meta-analyses of genome-wide association studies (meta-GWASs) and candidate gene studies have identified genetic variants associated with cardiovascular diseases, metabolic diseases and mood disorders. Although previous efforts were successful for individual disease conditions (single disease), limited information exists on shared genetic risk between these disorders. This article presents a detailed review and analysis of cardiometabolic diseases risk (CMD-R) genes that are also associated with mood disorders. First, we reviewed meta-GWASs published until January 2016, for the diseases 'type 2 diabetes, coronary artery disease, hypertension' and/or for the risk factors 'blood pressure, obesity, plasma lipid levels, insulin and glucose related traits'. We then searched the literature for published associations of these CMD-R genes with mood disorders. We considered studies that reported a significant association of at least one of the CMD-R genes and 'depression' or 'depressive disorder' or 'depressive symptoms' or 'bipolar disorder' or 'lithium treatment response in bipolar disorder', or 'serotonin reuptake inhibitors treatment response in major depression'. Our review revealed 24 potential pleiotropic genes that are likely to be shared between mood disorders and CMD-Rs. These genes include MTHFR, CACNA1D, CACNB2, GNAS, ADRB1, NCAN, REST, FTO, POMC, BDNF, CREB, ITIH4, LEP, GSK3B, SLC18A1, TLR4, PPP1R1B, APOE, CRY2, HTR1A, ADRA2A, TCF7L2, MTNR1B and IGF1. A pathway analysis of these genes revealed significant pathways: corticotrophin-releasing hormone signaling, AMPK signaling, cAMP-mediated or G-protein coupled receptor signaling, axonal guidance signaling, serotonin or dopamine receptors signaling, dopamine-DARPP32 feedback in cAMP signaling, circadian rhythm signaling and leptin signaling. Our review provides insights into the shared biological mechanisms of mood disorders and cardiometabolic diseases.
Collapse
Affiliation(s)
- A T Amare
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - K O Schubert
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, SA, Australia,Northern Adelaide Local Health Network, Mental Health Services, Adelaide, SA, Australia
| | - M Klingler-Hoffmann
- Adelaide Proteomics Centre, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - S Cohen-Woods
- School of Psychology, Faculty of Social and Behavioural Sciences, Flinders University, Adelaide, SA, Australia
| | - B T Baune
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, SA, Australia,Discipline of Psychiatry, School of Medicine, The University of Adelaide, North Terrace, Adelaide, SA 5005, Australia. E-mail:
| |
Collapse
|
39
|
Linden M, Helmbold K, Kempf J, Sippas S, Filss C, Langen KJ, Eisert A, Zepf FD. Dietary tryptophan depletion in humans using a simplified two amino acid formula - a pilot study. Food Nutr Res 2016; 60:29272. [PMID: 27989269 PMCID: PMC5165058 DOI: 10.3402/fnr.v60.29272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Revised: 09/26/2016] [Accepted: 11/03/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Acute tryptophan depletion (ATD) is a well-established dietary method in translational brain research used to briefly lower central nervous serotonin (5-hydroxytryptamine (5-HT)) synthesis. A simplified two amino acid ATD formula (ATDPHE/LEU) was developed while reducing the overall amount of amino acids (AAs), with the objective of administration especially in children and adolescents in future studies. OBJECTIVE This study investigated tryptophan (TRP) influx rates across the blood-brain barrier (BBB) after dietary ATDPHE/LEU administration relative to the ATD Moja-De protocol that has been established for use in children and adolescents. DESIGN Seventy-two healthy adults (50% females) were randomized into four groups and administered ATD Moja-De, its TRP-balanced control condition (BAL), ATDPHE/LEU, or its respective control mixture (BALPHE/LEU) in a counterbalanced, double-blind, between-subjects design. Blood samples were collected at baseline and at hourly intervals for 6 h after AA intake. Questionnaires about mood, taste, and challenge tolerance were completed at fixed time points. RESULTS Both challenge mixtures significantly reduced central nervous TRP influx as calculated by Michaelis-Menten kinetics relative to baseline and the respective control conditions with only mild and comparable side effects. A greater decline in TRP influx over the BBB after ATDPHE/LEU administration when compared with ATD Moja-De was detected without group effects for taste, challenge tolerance, and mood. There was unintended initial short increase in plasma TRP concentrations observed after ATDPHE/LEU intake, and a possible redistribution between free and protein-bound TRP triggered by protein synthesis stimulated by the ingested AAs may account for this finding. Moreover, a decline in TRP influx after BALPHE/LEU administration over a 6-h period was observed, and the large amount of PHE in the BALPHE/LEU mixture may be a possible explanation for this particular phenomenon, which could have led to an unexpected increase in displacement of TRP at the BBB in this control condition. CONCLUSIONS This pilot study provides preliminary evidence for the possibility of lowering TRP influx as calculated by Michaelis-Menten kinetics into the brain by using a simplified ATD protocol in humans. The simplified composition of only two AAs, the lower overall AA amount, and the appropriate tolerance are characteristics of the newly developed ATDPHE/LEU protocol. Future studies focusing on the effects of the ATDPHE/LEU protocol and its respective control condition on CSF 5-HIAA concentrations, as well as neurochemical studies in rodents, are needed to further validate this newly developed AA mixture before definite conclusions about its usability in ATD-related research in humans, its specificity, and additional effects can be made.
Collapse
Affiliation(s)
- Maike Linden
- Clinic for Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, Aachen, Germany.,JARA Translational Brain Medicine, Jülich Aachen Research Alliance, Jülich, Germany
| | - Katrin Helmbold
- Clinic for Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, Aachen, Germany.,JARA Translational Brain Medicine, Jülich Aachen Research Alliance, Jülich, Germany
| | - Janina Kempf
- Clinic for Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, Aachen, Germany.,JARA Translational Brain Medicine, Jülich Aachen Research Alliance, Jülich, Germany.,Faculty of Arts and Social Sciences, Maastricht University, Maastricht, The Netherlands
| | - Shabnam Sippas
- Clinic for Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, Aachen, Germany.,JARA Translational Brain Medicine, Jülich Aachen Research Alliance, Jülich, Germany.,Faculty of Arts and Social Sciences, Maastricht University, Maastricht, The Netherlands
| | - Christian Filss
- Section JARA-Brain, Jülich-Aachen Research Alliance (JARA), Jülich, Germany
| | - Karl-Josef Langen
- Section JARA-Brain, Jülich-Aachen Research Alliance (JARA), Jülich, Germany.,Institute of Neuroscience and Medicine (INM-3, -4, -5), Research Centre Jülich, Jülich, Germany.,Department of Nuclear Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Albrecht Eisert
- Department of Pharmacy, RWTH Aachen University, Aachen, Germany
| | - Florian Daniel Zepf
- Clinic for Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, Aachen, Germany.,JARA Translational Brain Medicine, Jülich Aachen Research Alliance, Jülich, Germany.,Centre and Discipline of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, School of Psychiatry and Clinical Neurosciences & School of Paediatrics and Child Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia, Perth, Australia.,Specialised Child and Adolescent Mental Health Services (CAMHS), Department of Health in Western Australia, Perth, Australia;
| |
Collapse
|
40
|
Luckhart C, Philippe TJ, Le François B, Vahid-Ansari F, Geddes SD, Béïque JC, Lagace DC, Daigle M, Albert PR. Sex-dependent adaptive changes in serotonin-1A autoreceptor function and anxiety in Deaf1-deficient mice. Mol Brain 2016; 9:77. [PMID: 27488351 PMCID: PMC4973060 DOI: 10.1186/s13041-016-0254-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 07/23/2016] [Indexed: 12/20/2022] Open
Abstract
The C (-1019) G rs6295 promoter polymorphism of the serotonin-1A (5-HT1A) receptor gene is associated with major depression in several but not all studies, suggesting that compensatory mechanisms mediate resilience. The rs6295 risk allele prevents binding of the repressor Deaf1 increasing 5-HT1A receptor gene transcription, and the Deaf1-/- mouse model shows an increase in 5-HT1A autoreceptor expression. In this study, Deaf1-/- mice bred on a mixed C57BL6-BALB/c background were compared to wild-type littermates for 5-HT1A autoreceptor function and behavior in males and females. Despite a sustained increase in 5-HT1A autoreceptor binding levels, the amplitude of the 5-HT1A autoreceptor-mediated current in 5-HT neurons was unaltered in Deaf1-/- mice, suggesting compensatory changes in receptor function. Consistent with increased 5-HT1A autoreceptor function in vivo, hypothermia induced by the 5-HT1A agonist DPAT was augmented in early generation male but not female Deaf1-/- mice, but was reduced with succeeding generations. Loss of Deaf1 resulted in a mild anxiety phenotype that was sex-and test-dependent, with no change in depression-like behavior. Male Deaf1 knockout mice displayed anxiety-like behavior in the open field and light-dark tests, while female Deaf1-/- mice showed increased anxiety only in the elevated plus maze. These data show that altered 5-HT1A autoreceptor regulation in male Deaf1-/- mice can be compensated for by generational adaptation of receptor response that may help to normalize behavior. The sex dependence of Deaf1 function in mice is consistent with a greater role for 5-HT1A autoreceptors in sensitivity to depression in men.
Collapse
Affiliation(s)
- Christine Luckhart
- Ottawa Hospital Research Institute (Neuroscience), Ottawa, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H-8M5, Canada
| | - Tristan J Philippe
- Ottawa Hospital Research Institute (Neuroscience), Ottawa, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H-8M5, Canada
| | - Brice Le François
- Ottawa Hospital Research Institute (Neuroscience), Ottawa, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H-8M5, Canada
| | - Faranak Vahid-Ansari
- Ottawa Hospital Research Institute (Neuroscience), Ottawa, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H-8M5, Canada
| | - Sean D Geddes
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H-8M5, Canada
| | - Jean-Claude Béïque
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H-8M5, Canada
| | - Diane C Lagace
- Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H-8M5, Canada
| | - Mireille Daigle
- Ottawa Hospital Research Institute (Neuroscience), Ottawa, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience), Ottawa, Canada. .,Department of Cellular and Molecular Medicine, University of Ottawa Brain and Mind Research Institute, 451 Smyth Road, Ottawa, ON, K1H-8M5, Canada.
| |
Collapse
|
41
|
Pantoja C, Hoagland A, Carroll EC, Karalis V, Conner A, Isacoff EY. Neuromodulatory Regulation of Behavioral Individuality in Zebrafish. Neuron 2016; 91:587-601. [PMID: 27397519 DOI: 10.1016/j.neuron.2016.06.016] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 05/05/2016] [Accepted: 06/08/2016] [Indexed: 11/30/2022]
Abstract
Inter-individual behavioral variation is thought to increase fitness and aid adaptation to environmental change, but the underlying mechanisms are poorly understood. We find that variation between individuals in neuromodulatory input contributes to individuality in short-term habituation of the zebrafish (Danio Rerio) acoustic startle response (ASR). ASR habituation varies greatly between individuals, but differences are stable over days and are heritable. Acoustic stimuli that activate ASR-command Mauthner cells also activate dorsal raphe nucleus (DRN) serotonergic neurons, which project to the vicinity of the Mauthner cells and their inputs. DRN neuron activity decreases during habituation in proportion to habituation and a genetic manipulation that reduces serotonin content in DRN neurons increases habituation, whereas serotonergic agonism or DRN activation with ChR2 reduces habituation. Finally, level of rundown of DRN activity co-segregates with extent of behavioral habituation across generations. Thus, variation between individuals in neuromodulatory input contributes to individuality in a core adaptive behavior. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Carlos Pantoja
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Adam Hoagland
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Elizabeth C Carroll
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Vasiliki Karalis
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Alden Conner
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Ehud Y Isacoff
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA; Bioscience Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
42
|
Scaini G, Rezin GT, Carvalho AF, Streck EL, Berk M, Quevedo J. Mitochondrial dysfunction in bipolar disorder: Evidence, pathophysiology and translational implications. Neurosci Biobehav Rev 2016; 68:694-713. [PMID: 27377693 DOI: 10.1016/j.neubiorev.2016.06.040] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 06/26/2016] [Accepted: 06/30/2016] [Indexed: 01/05/2023]
Abstract
Bipolar disorder (BD) is a chronic psychiatric illness characterized by severe and biphasic changes in mood. Several pathophysiological mechanisms have been hypothesized to underpin the neurobiology of BD, including the presence of mitochondrial dysfunction. A confluence of evidence points to an underlying dysfunction of mitochondria, including decreases in mitochondrial respiration, high-energy phosphates and pH; changes in mitochondrial morphology; increases in mitochondrial DNA polymorphisms; and downregulation of nuclear mRNA molecules and proteins involved in mitochondrial respiration. Mitochondria play a pivotal role in neuronal cell survival or death as regulators of both energy metabolism and cell survival and death pathways. Thus, in this review, we discuss the genetic and physiological components of mitochondria and the evidence for mitochondrial abnormalities in BD. The final part of this review discusses mitochondria as a potential target of therapeutic interventions in BD.
Collapse
Affiliation(s)
- Giselli Scaini
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Laboratory of Bioenergetics, Graduate Program in Health Sciences, Health Sciences Unit, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Gislaine T Rezin
- Laboratory of Clinical and Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Sul de Santa Catarina, Tubarão, SC, Brazil
| | - Andre F Carvalho
- Translational Psychiatry Research Group and Department of Clinical Medicine, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Emilio L Streck
- Laboratory of Bioenergetics, Graduate Program in Health Sciences, Health Sciences Unit, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, School of Medicine, Faculty of Health, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health and The Centre for Youth Mental Health, The Department of Psychiatry and The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - João Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA; Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil.
| |
Collapse
|
43
|
Miller MW, Sperbeck E, Robinson ME, Sadeh N, Wolf EJ, Hayes JP, Logue M, Schichman SA, Stone A, Milberg W, McGlinchey R. 5-HT2A Gene Variants Moderate the Association between PTSD and Reduced Default Mode Network Connectivity. Front Neurosci 2016; 10:299. [PMID: 27445670 PMCID: PMC4923242 DOI: 10.3389/fnins.2016.00299] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/13/2016] [Indexed: 02/02/2023] Open
Abstract
The default mode network (DMN) has been used to study disruptions of functional connectivity in a wide variety of psychiatric and neurological conditions, including posttraumatic stress disorder (PTSD). Studies indicate that the serotonin system exerts a modulatory influence on DMN connectivity; however, no prior study has examined associations between serotonin receptor gene variants and DMN connectivity in either clinical or healthy samples. We examined serotonin receptor single nucleotide polymorphisms (SNPs), PTSD, and their interactions for association with DMN connectivity in 134 White non-Hispanic veterans. We began by analyzing candidate SNPs identified in prior meta-analyses of relevant psychiatric traits and found that rs7997012 (an HTR2A SNP), implicated previously in anti-depressant medication response in the Sequenced Treatment Alternatives for Depression study (STAR(*)D; McMahon et al., 2006), interacted with PTSD to predict reduced connectivity between the posterior cingulate cortex (PCC) and the right medial prefrontal cortex and right middle temporal gyrus (MTG). rs130058 (HTR1B) was associated with connectivity between the PCC and right angular gyrus. We then expanded our analysis to 99 HTR1B and HTR2A SNPs and found two HTR2A SNPs (rs977003 and rs7322347) that significantly moderated the association between PTSD severity and the PCC-right MTG component of the DMN after correcting for multiple testing. Finally, to obtain a more precise localization of the most significant SNP × PTSD interaction, we performed a whole cortex vertex-wise analysis of the rs977003 effect. This analysis revealed the locus of the pre-frontal effect to be in portions of the superior frontal gyrus, while the temporal lobe effect was centered in the middle and inferior temporal gyri. These findings point to the influence of HTR2A variants on DMN connectivity and advance knowledge of the role of 5-HT2A receptors in the neurobiology of PTSD.
Collapse
Affiliation(s)
- Mark W Miller
- Behavioral Science Division, National Center for PTSD, VA Boston Healthcare SystemBoston, MA, USA; Department of Psychiatry, Boston University School of MedicineBoston, MA, USA
| | - Emily Sperbeck
- Department of Psychiatry, Boston University School of Medicine Boston, MA, USA
| | - Meghan E Robinson
- Neuroimaging Research for Veterans Center, VA Boston Healthcare SystemBoston, MA, USA; Geriatric Research Educational and Clinical Center and Translational Research Center for TBI and Stress Disorders, VA Boston Healthcare SystemBoston, MA, USA; Department of Neurology, Boston University School of MedicineBoston, MA, USA
| | - Naomi Sadeh
- Behavioral Science Division, National Center for PTSD, VA Boston Healthcare SystemBoston, MA, USA; Department of Psychiatry, Boston University School of MedicineBoston, MA, USA
| | - Erika J Wolf
- Behavioral Science Division, National Center for PTSD, VA Boston Healthcare SystemBoston, MA, USA; Department of Psychiatry, Boston University School of MedicineBoston, MA, USA
| | - Jasmeet P Hayes
- Behavioral Science Division, National Center for PTSD, VA Boston Healthcare SystemBoston, MA, USA; Department of Psychiatry, Boston University School of MedicineBoston, MA, USA
| | - Mark Logue
- Behavioral Science Division, National Center for PTSD, VA Boston Healthcare SystemBoston, MA, USA; Department of Psychiatry, Boston University School of MedicineBoston, MA, USA; Biomedical Genetics, Boston University School of MedicineBoston, MA, USA; Department of Biostatistics, Boston University School of Public HealthBoston, MA, USA
| | - Steven A Schichman
- Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System Little Rock, AR, USA
| | - Angie Stone
- Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System Little Rock, AR, USA
| | - William Milberg
- Geriatric Research Educational and Clinical Center and Translational Research Center for TBI and Stress Disorders, VA Boston Healthcare SystemBoston, MA, USA; Department of Psychiatry, Harvard Medical SchoolBoston, MA, USA
| | - Regina McGlinchey
- Geriatric Research Educational and Clinical Center and Translational Research Center for TBI and Stress Disorders, VA Boston Healthcare SystemBoston, MA, USA; Department of Psychiatry, Harvard Medical SchoolBoston, MA, USA
| |
Collapse
|
44
|
Cancelier K, Gomes LM, Carvalho-Silva M, Teixeira LJ, Rebelo J, Mota IT, Arent CO, Mariot E, Kist LW, Bogo MR, Quevedo J, Scaini G, Streck EL. Omega-3 Fatty Acids and Mood Stabilizers Alter Behavioural and Energy Metabolism Parameters in Animals Subjected to an Animal Model of Mania Induced by Fenproporex. Mol Neurobiol 2016; 54:3935-3947. [PMID: 27246566 DOI: 10.1007/s12035-016-9933-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/10/2016] [Indexed: 11/28/2022]
Abstract
Studies have shown that changes in energy metabolism are involved in the pathophysiology of bipolar disorder (BD). It was suggested that omega-3 (ω3) fatty acids have beneficial properties in the central nervous system and that this fatty acid plays an important role in energy metabolism. Therefore, the study aimed to evaluate the effect of ω3 fatty acids alone and in combination with lithium (Li) or valproate (VPA) on behaviour and parameters of energy metabolism in an animal model of mania induced by fenproporex. Our results showed that co-administration of ω3 fatty acids and Li was able to prevent and reverse the increase in locomotor and exploratory activity induced by fenproporex. The combination of ω3 fatty acids with VPA was only able to prevent the fenproporex-induced hyperactivity. For the energy metabolism parameters, our results showed that the administration of Fen for the reversal or prevention protocol inhibited the activities of succinate dehydrogenase, complex II and complex IV in the hippocampus. However, hippocampal creatine kinase (CK) activity was decreased only for the reversal protocol. The ω3 fatty acids, alone and in combination with VPA or Li, prevented and reversed the decrease in complex II, IV and succinate dehydrogenase activity, whereas the decrease in CK activity was only reversed after the co-administration of ω3 fatty acids and VPA. In conclusion, our results showed that the ω3 fatty acids combined with VPA or Li were able to prevent and reverse manic-like hyperactivity and the inhibition of energy metabolism in the hippocampus, suggesting that ω3 fatty acids may play an important role in the modulation of behavioural parameters and energy metabolism.
Collapse
Affiliation(s)
- Kizzy Cancelier
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Lara M Gomes
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Milena Carvalho-Silva
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Letícia J Teixeira
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Joyce Rebelo
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Isabella T Mota
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Camila O Arent
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Edemilson Mariot
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Luiza W Kist
- Laboratório de Biologia Genômica e Molecular, Departamento de Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Maurício R Bogo
- Laboratório de Biologia Genômica e Molecular, Departamento de Biologia Celular e Molecular, Faculdade de Biociências, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - João Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center (UTHealth), 1941 East Road, Ste. 5102, Houston, TX, USA.,Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center (UTHealth), Houston, TX, USA.,Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Giselli Scaini
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil. .,Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center (UTHealth), 1941 East Road, Ste. 5102, Houston, TX, USA.
| | - Emilio L Streck
- Laboratório de Bioenergética, Programa de Pós-graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| |
Collapse
|
45
|
Kaufman J, DeLorenzo C, Choudhury S, Parsey RV. The 5-HT1A receptor in Major Depressive Disorder. Eur Neuropsychopharmacol 2016; 26:397-410. [PMID: 26851834 PMCID: PMC5192019 DOI: 10.1016/j.euroneuro.2015.12.039] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 12/28/2015] [Accepted: 12/29/2015] [Indexed: 02/07/2023]
Abstract
Major Depressive Disorder (MDD) is a highly prevalent psychiatric diagnosis that is associated with a high degree of morbidity and mortality. This debilitating disorder is currently one of the leading causes of disability nationwide and is predicted to be the leading cause of disease burden by the year 2030. A large body of previous research has theorized that serotonergic dysfunction, specifically of the serotonin (5-HT) 1A receptor, plays a key role in the development of MDD. The purpose of this review is to describe the evolution of our current understanding of the serotonin 1A (5-HT1A) receptor and its role in the pathophysiology MDD through the discussion of animal, post-mortem, positron emission tomography (PET), pharmacologic and genetic studies.
Collapse
Affiliation(s)
- Joshua Kaufman
- Stony Brook University, Stony Brook, NY 11794, United States.
| | | | - Sunia Choudhury
- Stony Brook University, Stony Brook, NY 11794, United States
| | - Ramin V Parsey
- Stony Brook University, Stony Brook, NY 11794, United States
| |
Collapse
|
46
|
Donaldson ZR, le Francois B, Santos TL, Almli LM, Boldrini M, Champagne FA, Arango V, Mann JJ, Stockmeier CA, Galfalvy H, Albert PR, Ressler KJ, Hen R. The functional serotonin 1a receptor promoter polymorphism, rs6295, is associated with psychiatric illness and differences in transcription. Transl Psychiatry 2016; 6:e746. [PMID: 26926882 PMCID: PMC4872437 DOI: 10.1038/tp.2015.226] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 12/05/2015] [Indexed: 12/12/2022] Open
Abstract
The G/C single-nucleotide polymorphism in the serotonin 1a receptor promoter, rs6295, has previously been linked with depression, suicide and antidepressant responsiveness. In vitro studies suggest that rs6295 may have functional effects on the expression of the serotonin 1a receptor gene (HTR1A) through altered binding of a number of transcription factors. To further explore the relationship between rs6295, mental illness and gene expression, we performed dual epidemiological and biological studies. First, we genotyped a cohort of 1412 individuals, randomly split into discovery and replication cohorts, to examine the relationship between rs6295 and five psychiatric outcomes: history of psychiatric hospitalization, history of suicide attempts, history of substance or alcohol abuse, current posttraumatic stress disorder (PTSD), current depression. We found that the rs6295G allele is associated with increased risk for substance abuse, psychiatric hospitalization and suicide attempts. Overall, exposure to either childhood or non-childhood trauma resulted in increased risk for all psychiatric outcomes, but we did not observe a significant interaction between rs6295 and trauma in modulating psychiatric outcomes. In conjunction, we also investigated the potential impact of rs6295 on HTR1A expression in postmortem human brain tissue using relative allelic expression assays. We found more mRNA produced from the C versus the G-allele of rs6295 in the prefrontal cortex (PFC), but not in the midbrain of nonpsychiatric control subjects. Further, in the fetal cortex, rs6295C allele exhibited increased relative expression as early as gestational week 18 in humans. Finally, we found that the C:G allelic expression ratio was significantly neutralized in the PFC of subjects with major depressive disorder (MDD) who committed suicide as compared with controls, indicating that normal patterns of transcription may be disrupted in MDD/suicide. These data provide a putative biological mechanism underlying the association between rs6295, trauma and mental illness. Moreover, our results suggest that rs6295 may affect transcription during both gestational development and adulthood in a region-specific manner, acting as a risk factor for psychiatric illness. These findings provide a critical framework for conceptualizing the effects of a common functional genetic variant, trauma exposure and their impact on mental health.
Collapse
Affiliation(s)
- Z R Donaldson
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - B le Francois
- Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - T L Santos
- New York State Psychiatric Institute, New York, NY, USA
| | - L M Almli
- Department of Psychiatry, Emory University, Atlanta, GA, USA
| | - M Boldrini
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - F A Champagne
- Department of Psychology, Columbia University, New York, NY, USA
| | - V Arango
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - J J Mann
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - C A Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, USA
| | - H Galfalvy
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| | - P R Albert
- Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - K J Ressler
- Department of Psychiatry, Emory University, Atlanta, GA, USA
| | - R Hen
- New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
| |
Collapse
|
47
|
Eun TK, Jeong SH, Lee KY, Kim SH, Ahn YM, Bang YW, Joo EJ. Association between the 5-HTTLPR Genotype and Childhood Characteristics in Mood Disorders. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2016; 14:88-95. [PMID: 26792045 PMCID: PMC4730929 DOI: 10.9758/cpn.2016.14.1.88] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/11/2015] [Accepted: 11/17/2015] [Indexed: 12/31/2022]
Abstract
Objective The features of childhood attention deficit hyperactivity disorder (ADHD) are significantly associated with adult mood disorders. Some genetic factors may be common to both ADHD and mood disorders underlie the association between these two phenotypes. The present study aimed to determine whether a genetic role may be played by the serotonin transporter-linked polymorphic region (5-HTTLPR) in the childhood ADHD features of adult patients with mood disorders. Methods The present study included 232 patients with major depressive disorder (MDD), 154 patients with bipolar disorder (BPD), and 1,288 normal controls. Childhood ADHD features were assessed with the Korean version of the Wender Utah Rating Scale (WURS-K). The total score and the scores of three factors (impulsivity, inattention, and mood instability) from the WURS-K were analyzed to determine whether they were associated with the 5-HTTLPR genotype. Results In the BPD type II group, the 5-HTTLPR genotype was significantly associated with the total score (p=0.029) and the impulsivity factor (p=0.004) on the WURS-K. However, the inattention and mood instability factors were not associated with the 5-HTTLPR genotype. BPD type I, MDD and normal control groups did not exhibit any significant associations between the WURS-K scores and the 5-HTTLPR genotype. Conclusion The findings suggest that the 5-HTTLPR genotype may play a role in the impulsivity component of childhood ADHD in patients with BPD type II. Because of a small sample size and a single candidate gene, further studies investigating other candidate genes using a larger sample are warranted to determine any common genetic links.
Collapse
Affiliation(s)
- Tae Kyung Eun
- Department of Psychiatry, Eulji General Hospital, Seoul, Korea
| | - Seong Hoon Jeong
- Department of Neuropsychiatry, Eulji University School of Medicine, Daejeon, Korea.,Department of Psychiatry, Eulji University Hospital, Daejeon, Korea
| | - Kyu Young Lee
- Department of Psychiatry, Eulji General Hospital, Seoul, Korea.,Department of Neuropsychiatry, Eulji University School of Medicine, Daejeon, Korea
| | - Se Hyun Kim
- Department of Neuropsychiatry, Dongguk University Ilsan Hospital, Dongguk University School of Medicine, Goyang, Korea
| | - Yong Min Ahn
- Departments of Psychiatry and Behavioral Science, Seoul National University College of Medicine, Seoul, Korea.,Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Korea.,Department of Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | | | - Eun-Jeong Joo
- Department of Psychiatry, Eulji General Hospital, Seoul, Korea.,Department of Neuropsychiatry, Eulji University School of Medicine, Daejeon, Korea
| |
Collapse
|
48
|
Perroud N, Zewdie S, Stenz L, Adouan W, Bavamian S, Prada P, Nicastro R, Hasler R, Nallet A, Piguet C, Paoloni-Giacobino A, Aubry JM, Dayer A. METHYLATION OF SEROTONIN RECEPTOR 3A IN ADHD, BORDERLINE PERSONALITY, AND BIPOLAR DISORDERS: LINK WITH SEVERITY OF THE DISORDERS AND CHILDHOOD MALTREATMENT. Depress Anxiety 2016; 33:45-55. [PMID: 26350166 DOI: 10.1002/da.22406] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/28/2015] [Accepted: 08/02/2015] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Serotonin 3A receptor (5-HT3A R) is associated at the genetic and epigenetic levels with a variety of psychiatric disorders and interacts with early-life stress such as childhood maltreatment. We studied the impact of childhood maltreatment on the methylation status of the 5-HT3A R and its association with clinical severity outcomes in relation with a functional genetic polymorphism. METHODS Clinical severity indexes of 346 bipolar, borderline personality, and adult attention deficit hyperactivity disorders patients were tested for association with the DNA methylation status of eight 5-HT3A R gene CpGs. Relationship between the functional variant rs1062613 (C > T) and methylation status on severity of the disorders were also assessed. RESULTS Childhood maltreatment was associated with higher severity of the disease (higher number of mood episodes, history of suicide attempts, hospitalization, and younger age at onset) across disorders and within each individual disorder. This effect was mediated by two 5-HT3A R CpGs. Compared to T allele carriers, CC carriers had higher methylation status at one CpG located 1 bp upstream of this variant. CONCLUSIONS This study shows that epigenetic modification of the 5-HT3A R is involved in the mechanism underlying the relationship between maltreatment in childhood and the severity of several psychiatric disorders in adulthood.
Collapse
Affiliation(s)
- Nader Perroud
- Department of Mental Health and Psychiatry, Service of Psychiatric Specialties, University Hospitals of Geneva, Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | | | - Ludwig Stenz
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.,Department of Basic Neuroscience, University of Geneva, Switzerland
| | - Wafae Adouan
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Sabine Bavamian
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.,Department of Basic Neuroscience, University of Geneva, Switzerland
| | - Paco Prada
- Department of Mental Health and Psychiatry, Service of Psychiatric Specialties, University Hospitals of Geneva, Geneva, Switzerland
| | - Rosetta Nicastro
- Department of Mental Health and Psychiatry, Service of Psychiatric Specialties, University Hospitals of Geneva, Geneva, Switzerland
| | - Roland Hasler
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Audrey Nallet
- Department of Mental Health and Psychiatry, Service of Psychiatric Specialties, University Hospitals of Geneva, Geneva, Switzerland
| | - Camille Piguet
- Department of Mental Health and Psychiatry, Service of Psychiatric Specialties, University Hospitals of Geneva, Geneva, Switzerland
| | - Ariane Paoloni-Giacobino
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Jean-Michel Aubry
- Department of Mental Health and Psychiatry, Service of Psychiatric Specialties, University Hospitals of Geneva, Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Alexandre Dayer
- Department of Mental Health and Psychiatry, Service of Psychiatric Specialties, University Hospitals of Geneva, Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Geneva, Switzerland.,Department of Basic Neuroscience, University of Geneva, Switzerland
| |
Collapse
|
49
|
A review of genetic alterations in the serotonin pathway and their correlation with psychotic diseases and response to atypical antipsychotics. Schizophr Res 2016; 170:18-29. [PMID: 26644303 DOI: 10.1016/j.schres.2015.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 10/30/2015] [Accepted: 11/02/2015] [Indexed: 02/06/2023]
Abstract
Serotonin is a neurotransmitter that plays a predominant role in mood regulation. The importance of the serotonin pathway in controlling behavior and mental status is well recognized. All the serotonin elements - serotonin receptors, serotonin transporter, tryptophan hydroxylase and monoamine oxidase proteins - can show alterations in terms of mRNA or protein levels and protein sequence, in schizophrenia and bipolar disorder. Additionally, when examining the genes sequences of all serotonin elements, several single nucleotide polymorphisms (SNPs) have been found to be more prevalent in schizophrenic or bipolar patients than in healthy individuals. Several of these alterations have been associated either with different phenotypes between patients and healthy individuals or with the response of psychiatric patients to the treatment with atypical antipsychotics. The complex pattern of genetic diversity within the serotonin pathway hampers efforts to identify the key variations contributing to an individual's susceptibility to the disease. In this review article, we summarize all genetic alterations found across the serotonin pathway, we provide information on whether and how they affect schizophrenia or bipolar disorder phenotypes, and, on the contribution of familial relationships on their detection frequencies. Furthermore, we provide evidence on whether and how specific gene polymorphisms affect the outcome of schizophrenic or bipolar patients of different ethnic groups, in response to treatment with atypical antipsychotics. All data are discussed thoroughly, providing prospective for future studies.
Collapse
|
50
|
Smoller JW. The Genetics of Stress-Related Disorders: PTSD, Depression, and Anxiety Disorders. Neuropsychopharmacology 2016; 41:297-319. [PMID: 26321314 PMCID: PMC4677147 DOI: 10.1038/npp.2015.266] [Citation(s) in RCA: 281] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/05/2015] [Accepted: 08/26/2015] [Indexed: 02/06/2023]
Abstract
Research into the causes of psychopathology has largely focused on two broad etiologic factors: genetic vulnerability and environmental stressors. An important role for familial/heritable factors in the etiology of a broad range of psychiatric disorders was established well before the modern era of genomic research. This review focuses on the genetic basis of three disorder categories-posttraumatic stress disorder (PTSD), major depressive disorder (MDD), and the anxiety disorders-for which environmental stressors and stress responses are understood to be central to pathogenesis. Each of these disorders aggregates in families and is moderately heritable. More recently, molecular genetic approaches, including genome-wide studies of genetic variation, have been applied to identify specific risk variants. In this review, I summarize evidence for genetic contributions to PTSD, MDD, and the anxiety disorders including genetic epidemiology, the role of common genetic variation, the role of rare and structural variation, and the role of gene-environment interaction. Available data suggest that stress-related disorders are highly complex and polygenic and, despite substantial progress in other areas of psychiatric genetics, few risk loci have been identified for these disorders. Progress in this area will likely require analysis of much larger sample sizes than have been reported to date. The phenotypic complexity and genetic overlap among these disorders present further challenges. The review concludes with a discussion of prospects for clinical translation of genetic findings and future directions for research.
Collapse
Affiliation(s)
- Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|