1
|
Hu Y, Zou Y, Zhang M, Yan J, Zheng Y, Chen Y. The relationship between major depressive disorder and dementia: A bidirectional two-sample Mendelian randomization study. J Affect Disord 2024; 355:167-174. [PMID: 38548196 DOI: 10.1016/j.jad.2024.03.149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/07/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) and dementia psychiatric and neurological diseases that are clinically widespread, but whether there is a causal link between them is still unclear. In this study, bidirectional two-sample Mendelian randomization (MR) was used to investigate the potential causal relationship between MDD and dementia via a genome-wide association study (GWAS) database, containing samples from the European population. METHOD We collected data on MDD and common clinical dementia subtypes from GWAS, including Alzheimer's disease (AD), frontotemporal dementia (FTD), dementia with Lewy bodies (DLB), Parkinson's disease with dementia (PDD), and vascular dementia (VaD). A series of bidirectional two-sample MR studies and correlation sensitivity analysis were carried out. RESULTS In the study of the effect of MDD on dementia subtypes, no causal relationship was found between MDD and dementia subtypes other than VaD, inverse variance weighted (IVW) method: odds ratio (OR), 2.131; 95 % confidence interval (CI), 1.249-3.639, P = 0.006; MDD-AD: OR, 1.000; 95 % CI, 0.999-1.001, P = 0.537; MDD-FTD: OR, 1.476; 95 % CI, 0.471-4.627, P = 0.505; MDD-PDD: OR, 0.592; 95 % CI, 0.204-1.718, P = 0.335; MR-Egger method: MDD-DLB: OR, 0.582; 95 % CI, 0.021-15.962, P = 0.751. In reverse MR analyses, no dementia subtype was found to be a risk factor for MDD. LIMITATIONS The results of this study may not be generalizable to non-European populations. CONCLUSION MDD was identified as a potential risk factor for VaD, but no dementia subtype was found to be a risk factor for MDD. These results suggest a new avenue for the prevention of VaD.
Collapse
Affiliation(s)
- Yijun Hu
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China; Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, China
| | - Yuntao Zou
- Weifang Hospital of Traditional Chinese Medicine, Wei Fang, China
| | - Meng Zhang
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China; Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, China
| | - Jinglan Yan
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China; Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, China
| | - Yuanjia Zheng
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China; Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, China
| | - Yongjun Chen
- Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China; Innovation Research Institute of Chinese Medicine, Shandong University of Traditional Chinese Medicine, China; Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, PR China.
| |
Collapse
|
2
|
Poggi G, Klaus F, Pryce CR. Pathophysiology in cortico-amygdala circuits and excessive aversion processing: the role of oligodendrocytes and myelination. Brain Commun 2024; 6:fcae140. [PMID: 38712320 PMCID: PMC11073757 DOI: 10.1093/braincomms/fcae140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/27/2023] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
Stress-related psychiatric illnesses, such as major depressive disorder, anxiety and post-traumatic stress disorder, present with alterations in emotional processing, including excessive processing of negative/aversive stimuli and events. The bidirectional human/primate brain circuit comprising anterior cingulate cortex and amygdala is of fundamental importance in processing emotional stimuli, and in rodents the medial prefrontal cortex-amygdala circuit is to some extent analogous in structure and function. Here, we assess the comparative evidence for: (i) Anterior cingulate/medial prefrontal cortex<->amygdala bidirectional neural circuits as major contributors to aversive stimulus processing; (ii) Structural and functional changes in anterior cingulate cortex<->amygdala circuit associated with excessive aversion processing in stress-related neuropsychiatric disorders, and in medial prefrontal cortex<->amygdala circuit in rodent models of chronic stress-induced increased aversion reactivity; and (iii) Altered status of oligodendrocytes and their oligodendrocyte lineage cells and myelination in anterior cingulate/medial prefrontal cortex<->amygdala circuits in stress-related neuropsychiatric disorders and stress models. The comparative evidence from humans and rodents is that their respective anterior cingulate/medial prefrontal cortex<->amygdala circuits are integral to adaptive aversion processing. However, at the sub-regional level, the anterior cingulate/medial prefrontal cortex structure-function analogy is incomplete, and differences as well as similarities need to be taken into account. Structure-function imaging studies demonstrate that these neural circuits are altered in both human stress-related neuropsychiatric disorders and rodent models of stress-induced increased aversion processing. In both cases, the changes include altered white matter integrity, albeit the current evidence indicates that this is decreased in humans and increased in rodent models. At the cellular-molecular level, in both humans and rodents, the current evidence is that stress disorders do present with changes in oligodendrocyte lineage, oligodendrocytes and/or myelin in these neural circuits, but these changes are often discordant between and even within species. Nonetheless, by integrating the current comparative evidence, this review provides a timely insight into this field and should function to inform future studies-human, monkey and rodent-to ascertain whether or not the oligodendrocyte lineage and myelination are causally involved in the pathophysiology of stress-related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Giulia Poggi
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, CH-8008 Zurich, Switzerland
| | - Federica Klaus
- Department of Psychiatry, University of California San Diego, San Diego, CA 92093, USA
- Desert-Pacific Mental Illness Research Education and Clinical Center, VA San Diego Healthcare System, San Diego, CA 92093, USA
| | - Christopher R Pryce
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, CH-8008 Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
- URPP Adaptive Brain Circuits in Development and Learning (AdaBD), University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
3
|
Bhivandkar S, Sarfraz Z, Jain L, Bachu A, Malo PK, Hsu M, Ayub S, Poudel L, Kumar H, Loh H, Tazin F, Ahmed S, Suzuki J. Therapeutic Potential of Buprenorphine in Depression: A Meta-Analysis of Current Evidence. J Clin Med Res 2024; 16:46-55. [PMID: 38550549 PMCID: PMC10970042 DOI: 10.14740/jocmr5050] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/06/2024] [Indexed: 03/12/2025] Open
Abstract
BACKGROUND Emerging research indicates buprenorphine, used in management of opioid use disorder, has attracted interest for its potential in treating a variety of psychiatric conditions. This meta-analysis aimed to determine the efficacy of buprenorphine in treating symptoms of depression. METHODS Using Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines, a search was conducted of several databases until April 25, 2022, for English language articles related to buprenorphine and its use in treating various mental health conditions. Standardized mean differences (SMDs) and its 95% confidence intervals (CIs) were reported for the Hamilton Rating Scale for Depression (HAM-D) and the Montgomery-Asberg Depression Rating Scale (MADRS) scores. Statistical analyses were performed using Cochrane RevMan 5. RESULTS Of the 1,347 identified studies, six clinical trials were included. MADRS-10 least square mean difference (LSMD) inter-group assessment favored buprenorphine over placebo, but it lacked statistical significance. Similarly, MADRS scores as well as HAM-D inter-group assessment were in favor of buprenorphine, however, were not statistically significant. These findings suggest a potential therapeutic role for buprenorphine in treating depression, albeit with caution due to the observed lack of statistical significance and the potential for confounding factors. CONCLUSIONS Preliminary evidence suggests potential efficacy of buprenorphine at lower doses in improving improving outcomes specifically related to depression. However, due to limitations in statistical significance and possible confounding factors, entail cautious interpretation. Further rigorous research is needed to investigate the long-term effects, optimal dosing, and determine the role of adjuvant drug therapy.
Collapse
Affiliation(s)
- Siddhi Bhivandkar
- Department of Psychiatry, Boston University Medical Center/Boston University School of Medicine, Boston, MA, USA
| | - Zouina Sarfraz
- Department of Psychiatry, Fatima Jinnah Medical University, Lahore, Pakistan
| | - Lakshit Jain
- Department of Psychiatry, Connecticut Valley Hospital, Middletown, CT, USA
| | - Anil Bachu
- Department of Psychiatry, Baptist Health - UAMS Psychiatry Residency Program, North Little Rock, AR, USA
| | - Palash Kumar Malo
- Department of Psychiatry, Centre for Brain Research, Indian Institute of Science, Bengaluru, India
| | - Michael Hsu
- Department of Psychiatry, Emory University School of Medicine, Atlanta, GA, USA
| | - Shahana Ayub
- Institute of Living/Hartford Hospital Hartford, Hartford, CT, USA
| | - Laxmi Poudel
- Department of Psychiatry, Nepalgunj Medical College, Nepalgunj, Banke, Nepal
| | | | - Hanyou Loh
- National University of Singapore, Singapore, Singapore
| | - Faria Tazin
- Department of Psychiatry, East Liverpool City Hospital, East Liverpool, OH, USA
| | - Saeed Ahmed
- Saint Francis Hospital and Medical Center, Hartford, CT, USA
| | - Joji Suzuki
- Department of Psychiatry, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Molecular characterization of depression trait and state. Mol Psychiatry 2022; 27:1083-1094. [PMID: 34686766 DOI: 10.1038/s41380-021-01347-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 09/23/2021] [Accepted: 10/04/2021] [Indexed: 11/09/2022]
Abstract
Major depressive disorder (MDD) is a brain disorder often characterized by recurrent episode and remission phases. The molecular correlates of MDD have been investigated in case-control comparisons, but the biological alterations associated with illness trait (regardless of clinical phase) or current state (symptomatic and remitted phases) remain largely unknown, limiting targeted drug discovery. To characterize MDD trait- and state-dependent changes, in single or recurrent depressive episode or remission, we generated transcriptomic profiles of subgenual anterior cingulate cortex of postmortem subjects in first MDD episode (n = 20), in remission after a single episode (n = 15), in recurrent episode (n = 20), in remission after recurring episodes (n = 15) and control subject (n = 20). We analyzed the data at the gene, biological pathway, and cell-specific molecular levels, investigated putative causal events and therapeutic leads. MDD-trait was associated with genes involved in inflammation, immune activation, and reduced bioenergetics (q < 0.05) whereas MDD-states were associated with altered neuronal structure and reduced neurotransmission (q < 0.05). Cell-level deconvolution of transcriptomic data showed significant change in density of GABAergic interneurons positive for corticotropin-releasing hormone, somatostatin, or vasoactive-intestinal peptide (p < 3 × 10-3). A probabilistic Bayesian-network approach showed causal roles of immune-system-activation (q < 8.67 × 10-3), cytokine-response (q < 4.79 × 10-27) and oxidative-stress (q < 2.05 × 10-3) across MDD-phases. Gene-sets associated with these putative causal changes show inverse associations with the transcriptomic effects of dopaminergic and monoaminergic ligands. The study provides first insights into distinct cellular and molecular pathologies associated with trait- and state-MDD, on plasticity mechanisms linking the two pathologies, and on a method of drug discovery focused on putative disease-causing pathways.
Collapse
|
5
|
Saloner R, Cherner M, Grelotti DJ, Paolillo EW, Moore DJ, Heaton RK, Letendre SL, Kumar A, Grant I, Ellis RJ. Lower CSF homovanillic acid relates to higher burden of neuroinflammation and depression in people with HIV disease. Brain Behav Immun 2020; 90:353-363. [PMID: 32966871 PMCID: PMC7544671 DOI: 10.1016/j.bbi.2020.09.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/08/2020] [Accepted: 09/13/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND HIV-related neuroinflammation has been proposed as a catalyst for dopaminergic dysregulation in mesocortical pathways, which may contribute to the pathogenesis of depression. Abnormalities in dopaminergic neurotransmission and depression are common in people with HIV (PWH), however the link between dopamine (DA) and depression in PWH is poorly characterized. This study investigated CSF dopaminergic biomarkers, specifically DA and its metabolite, homovanillic acid (HVA), and examined their relationship with depressive symptoms and CSF neuroinflammatory markers in PWH and HIV-seronegative (HIV-) individuals. METHODS Participants were 102 HIV- individuals and 123 PWH (mean age = 42) who underwent neuropsychiatric evaluations and lumbar puncture. Current depression severity was classified using the Beck Depression Inventory-II (BDI-II). CSF was assayed for DA and HVA using high performance liquid chromatography and neuroinflammatory markers using immunoassays. Linear regressions modelled BDI-II scores as a function of HIV, dopaminergic biomarker z-scores, and their interaction, controlling for psychosocial factors. Correlational analyses examined dopaminergic and neuroinflammatory relationships. RESULTS PWH had significantly higher BDI-II scores than HIV- participants. DA and HVA were not associated with HIV status but both significantly moderated the effect of HIV on BDI-II scores, such that PWH exhibited higher depressive symptoms than HIV- participants only at lower concentrations of HVA (z ≤ 0.06) and DA (z ≤ 0.11). In PWH only, lower HVA significantly correlated with higher BDI-II scores and higher neuroinflammation, including higher MCP-1 and IP-10. CONCLUSIONS Results suggest that the pathophysiology of depression in PWH differs from that in HIV- individuals. Specifically, lower central dopaminergic activity was selectively associated with greater depressive symptoms and neuroinflammation in PWH. With the rise in consideration of DA agonists for the treatment of depression, these results suggest that PWH may show a greater response to these agents than their HIV- peers.
Collapse
Affiliation(s)
- Rowan Saloner
- San Diego State University/University of California, San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA; Department of Psychiatry, University of California, San Diego, HIV Neurobehavioral Research Program, San Diego, CA, USA.
| | - Mariana Cherner
- Department of Psychiatry, University of California, San Diego, HIV Neurobehavioral Research Program, San Diego, CA, USA
| | - David J Grelotti
- Department of Psychiatry, University of California, San Diego, HIV Neurobehavioral Research Program, San Diego, CA, USA
| | - Emily W Paolillo
- San Diego State University/University of California, San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA; Department of Psychiatry, University of California, San Diego, HIV Neurobehavioral Research Program, San Diego, CA, USA
| | - David J Moore
- Department of Psychiatry, University of California, San Diego, HIV Neurobehavioral Research Program, San Diego, CA, USA
| | - Robert K Heaton
- Department of Psychiatry, University of California, San Diego, HIV Neurobehavioral Research Program, San Diego, CA, USA
| | - Scott L Letendre
- Department of Psychiatry, University of California, San Diego, HIV Neurobehavioral Research Program, San Diego, CA, USA
| | - Adarsh Kumar
- Department of Psychiatry and Behavioral Sciences, University of Miami, Miami, FL, USA
| | - Igor Grant
- Department of Psychiatry, University of California, San Diego, HIV Neurobehavioral Research Program, San Diego, CA, USA
| | - Ronald J Ellis
- Department of Psychiatry, University of California, San Diego, HIV Neurobehavioral Research Program, San Diego, CA, USA; Department of Neurosciences, University of California, San Diego, San Diego, CA, USA
| |
Collapse
|
6
|
Yazdani A, Mendez-Giraldez R, Yazdani A, Kosorok MR, Roussos P. Differential gene regulatory pattern in the human brain from schizophrenia using transcriptomic-causal network. BMC Bioinformatics 2020; 21:469. [PMID: 33087039 PMCID: PMC7579819 DOI: 10.1186/s12859-020-03753-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Common and complex traits are the consequence of the interaction and regulation of multiple genes simultaneously, therefore characterizing the interconnectivity of genes is essential to unravel the underlying biological networks. However, the focus of many studies is on the differential expression of individual genes or on co-expression analysis. METHODS Going beyond analysis of one gene at a time, we systematically integrated transcriptomics, genotypes and Hi-C data to identify interconnectivities among individual genes as a causal network. We utilized different machine learning techniques to extract information from the network and identify differential regulatory pattern between cases and controls. We used data from the Allen Brain Atlas for replication. RESULTS Employing the integrative systems approach on the data from CommonMind Consortium showed that gene transcription is controlled by genetic variants proximal to the gene (cis-regulatory factors), and transcribed distal genes (trans-regulatory factors). We identified differential gene regulatory patterns in SCZ-cases versus controls and novel SCZ-associated genes that may play roles in the disorder since some of them are primary expressed in human brain. In addition, we observed genes known associated with SCZ are not likely (OR = 0.59) to have high impacts (degree > 3) on the network. CONCLUSIONS Causal networks could reveal underlying patterns and the role of genes individually and as a group. Establishing principles that govern relationships between genes provides a mechanistic understanding of the dysregulated gene transcription patterns in SCZ and creates more efficient experimental designs for further studies. This information cannot be obtained by studying a single gene at the time.
Collapse
Affiliation(s)
- Akram Yazdani
- Department of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, 120 Mason Farm Road, Genetic Medicine Building, CB#7361, Chapel Hill, NC, 27599-7264, USA.
| | - Raul Mendez-Giraldez
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Azam Yazdani
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael R Kosorok
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Panos Roussos
- Department of Psychiatry, Pamela Sklar Division of Psychiatric Genomics and Friedman Brain Institute, Icahn School of Medicine At Mount Sinai, Hess CSM Building Floor 9 Room 107, 1470 Madison Ave, New York, NY, 10029, USA.
- Mental Illness Research Education and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY, 10468, USA.
| |
Collapse
|
7
|
Ciobanu LG, Sachdev PS, Trollor JN, Reppermund S, Thalamuthu A, Mather KA, Cohen-Woods S, Stacey D, Toben C, Schubert KO, Baune BT. Downregulated transferrin receptor in the blood predicts recurrent MDD in the elderly cohort: A fuzzy forests approach. J Affect Disord 2020; 267:42-48. [PMID: 32063571 DOI: 10.1016/j.jad.2020.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/28/2020] [Accepted: 02/01/2020] [Indexed: 01/22/2023]
Abstract
BACKGROUND At present, no predictive markers for Major Depressive Disorder (MDD) exist. The search for such markers has been challenging due to clinical and molecular heterogeneity of MDD, the lack of statistical power in studies and suboptimal statistical tools applied to multidimensional data. Machine learning is a powerful approach to mitigate some of these limitations. METHODS We aimed to identify the predictive markers of recurrent MDD in the elderly using peripheral whole blood from the Sydney Memory and Aging Study (SMAS) (N = 521, aged over 65) and adopting machine learning methodology on transcriptome data. Fuzzy Forests is a Random Forests-based classification algorithm that takes advantage of the co-expression network structure between genes; it allows to alleviate the problem of p >> n via reducing the dimensionality of transcriptomic feature space. RESULTS By adopting Fuzzy Forests on transcriptome data, we found that the downregulated TFRC (transferrin receptor) can predict recurrent MDD with an accuracy of 63%. LIMITATIONS Although we corrected our data for several important confounders, we were not able to account for the comorbidities and medication taken, which may be numerous in the elderly and might have affected the levels of gene transcription. CONCLUSIONS We found that downregulated TFRC is predictive of recurrent MDD, which is consistent with the previous literature, indicating the role of the innate immune system in depression. This study is the first to successfully apply Fuzzy Forests methodology on psychiatric condition, opening, therefore, a methodological avenue that can lead to clinically useful predictive markers of complex traits.
Collapse
Affiliation(s)
- Liliana G Ciobanu
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, South Australia, Australia; Quality Use of Medicines and Pharmacy Research Centre, School of Pharmacy and Medical Sciences, University of South Australia, Australia
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, UNSW Sydney, New South Wales, Australia
| | - Julian N Trollor
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, UNSW Sydney, New South Wales, Australia; Department of Developmental Disability Neuropsychiatry, School of Psychiatry, UNSW Sydney, New South Wales, Australia
| | - Simone Reppermund
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, UNSW Sydney, New South Wales, Australia; Department of Developmental Disability Neuropsychiatry, School of Psychiatry, UNSW Sydney, New South Wales, Australia
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, UNSW Sydney, New South Wales, Australia
| | - Karen A Mather
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, UNSW Sydney, New South Wales, Australia; Neuroscience Research Australia, Randwick, Australia
| | - Sarah Cohen-Woods
- School of Psychology, Flinders University, South Australia, Australia
| | - David Stacey
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | - Catherine Toben
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, South Australia, Australia
| | - K Oliver Schubert
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, South Australia, Australia; Northern Adelaide Local Health Network, Mental Health Services, Lyell McEwin Hospital, Elizabeth Vale, South Australia, Australia
| | - Bernhard T Baune
- Department of Psychiatry and Psychotherapy, University of Münster, Münster, Germany; Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
8
|
Why context matters? Divisive normalization and canonical microcircuits in psychiatric disorders. Neurosci Res 2019; 156:130-140. [PMID: 31628970 DOI: 10.1016/j.neures.2019.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/30/2019] [Accepted: 09/04/2019] [Indexed: 11/20/2022]
Abstract
Neural activity on cellular, regional, and behavioral levels shows context-dependence. Here we suggest the processing of input-output relationships in terms divisive normalization (DN), including (i) summing/averaging inputs and (ii) normalizing output against input stages, as a computational mechanism to underlie context-dependence. Input summation and output normalization are mediated by input-output relationships in canonical microcircuits (CM). DN/CM are altered in psychiatric disorders like schizophrenia or depression whose various symptoms can be characterized by abnormal context-dependence.
Collapse
|
9
|
Serafini G, Santi F, Gonda X, Aguglia A, Fiorillo A, Pompili M, Carvalho AF, Amore M. Predictors of recurrence in a sample of 508 outpatients with major depressive disorder. J Psychiatr Res 2019; 114:80-87. [PMID: 31051436 DOI: 10.1016/j.jpsychires.2019.04.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Specific predictors of relapse/recurrence in major depressive disorder (MDD) have been identified but evidence across studies are inconsistent. This study aimed to identify the most relevant socio-demographic/clinical predictors of MDD recurrence in a sample of 508 outpatients. METHODS This naturalistic cohort study included 508 currently euthymic MDD patients (mean age = 54.1 ± 16.2) of which 53.9% had a single and 46.1% recurrent depressive episodes. A detailed data collection was performed and illness histories were retraced through clinical files and lifetime computerized medical records. RESULTS Compared to patients with single episode, MDD patients with recurrent episodes significantly differ regarding current age, gender, working status, positive history of psychiatric disorders in family, first-lifetime illness episode characteristics, first-episode and current psychotic symptoms, current melancholic features and seasonality, age at first treatment, duration of untreated illness, and comorbid cardiovascular/endocrinological conditions. However, after multivariate analyses controlling for current age, gender, educational level, working status differences, psychiatric conditions in family, and age of illness episode, recurrence was associated with older age (p ≤ .001), younger age at first treatment (p ≤ .005), being treated with previous psychoactive treatments (p .001), and longer duration of untreated illness (p .001). CONCLUSIONS The variables associated with MDD recurrence identified in the current study may aid in the stratification of patients who could benefit from more intensive maintenance treatments for MDD. However, clinicians should rapidly identify cases that are not likely to recur in order to avoid unnecessary treatments which are commonly considered as the standard of care.
Collapse
Affiliation(s)
- Gianluca Serafini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Francesca Santi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Xenia Gonda
- Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary; MTA-SE Neuropsychopharmacology and Neurochemistry Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary; NAP-2-SE New Antidepressant Target Research Group, Semmelweis University, Budapest, Hungary
| | - Andrea Aguglia
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Andrea Fiorillo
- Department of Psychiatry, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Maurizio Pompili
- Department of Neurosciences, Suicide Prevention Center, Sant'Andrea Hospital, University of Rome, Rome, Italy
| | - André F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Mario Amore
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
10
|
Association between Salivary Alpha-Amylase and Executive Functioning in Healthy Children. SPANISH JOURNAL OF PSYCHOLOGY 2019; 22:E24. [DOI: 10.1017/sjp.2019.26] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
AbstractThe main aim of this study was to confirm the relationship between executive performance and salivary alpha-amylase (SAA) activity in a sample of 64 healthy children (39 boys), and compare it to the association of SAA output and salivary flow rate (SFR). Executive functioning was assessed via fluency, trail-making, rings and inhibition tasks from the Batería de Evaluación Neuropsicológica de la Función Ejecutiva en Niños [Battery of Neuropsychological Assessment for Executive Function in Children] (ENFEN), merged into an ENFEN total score. SAA activity, output, and SFR were measured at baseline, one minute before, and one minute after the end of a neuropsychological testing session. Our results confirmed a direct, linear and significant association between SAA activity and executive functioning, r(64) = .351, p < .05, and extended it to SAA output, r(64) =.431, p < .05. The mean level of SAA output was the best predictor of executive functioning (β = .431, p < .05) and explained 18.2 % of the variance in ENFEN total score. In sum, and compared to SAA activity, measuring SAA output may be a more precise and indirect marker to assess executive functioning in children.
Collapse
|
11
|
Plasma microRNA expression levels and their targeted pathways in patients with major depressive disorder who are responsive to duloxetine treatment. J Psychiatr Res 2019; 110:38-44. [PMID: 30580082 DOI: 10.1016/j.jpsychires.2018.12.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/14/2018] [Accepted: 12/06/2018] [Indexed: 11/21/2022]
Abstract
Major depressive disorder (MDD) is a complex disorder with many pathways known to contribute to its pathogenesis, such as apoptotic signaling, with antidepressants having been shown to target these pathways. In this study, we explored microRNAs as predictive markers of drug response to duloxetine, a serotonin-norepinephrine reuptake inhibiter, using peripheral blood samples from 3 independent clinical trials (NCT00635219; NCT0059991; NCT01140906) comparing 6-8 weeks of treatment with duloxetine to placebo treatment in patients with MDD. Plasma microRNA was extracted and sequenced using the Ion Proton Sequencer. Rank feature selection analysis was used to identify microRNAs in the top 10th percentile for their differentiating ability between patients who remitted and did not remit with duloxetine treatment. The results were then compared between the 3 trials to see their replicability. To further validate our findings, we reasoned that the pathways targeted by these microRNAs would be those shown to be altered in MDD in pathway enrichment analysis. Hsa-miR-23a-3p, hsa-miR-16-5p, hsa-miR-146a-5p and hsa-miR-21-5p were identified in 2 or more trials as being able to differentiate patients who would remit with duloxetine treatment using samples collected before treatment initiation, suggesting that they may be good candidates for identification of predictive biomarkers of duloxetine response. Pathway enrichment analysis further showed that microRNAs identified as differentiating for duloxetine response target the apoptosis signaling pathway. Future studies examining these microRNAs outside of a clinical trial setting and exploring their role in MDD may further our understanding of MDD and antidepressant response.
Collapse
|
12
|
Serafini G, Vazquez GH, Gonda X, Pompili M, Rihmer Z, Amore M. Depressive residual symptoms are associated with illness course characteristics in a sample of outpatients with bipolar disorder. Eur Arch Psychiatry Clin Neurosci 2018; 268:757-768. [PMID: 29417206 DOI: 10.1007/s00406-018-0875-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 01/24/2018] [Indexed: 01/23/2023]
Abstract
Rates of 50-70% of residual symptoms referring to subsyndromal manifestations between episodes that do not meet the required criteria for episode definition were reported in bipolar disorder (BD). However, the specific role of these symptoms on the course of BD patients is poorly understood; thus, we aimed to investigate factors associated with depressive residual symptoms. Overall, 255 currently euthymic BD outpatients on maintenance treatment, including 95 (37.2%) males and 160 (62.8%) females, were consecutively recruited at the Section of Psychiatry, Department of Neuroscience, University of Genoa (Italy) and underwent detailed structured interviews, comprehensive clinical interviews, and clinical record reviews for assessment/collection of relevant information concerning the course of illness and clinical status including cross-referral of all available information. After categorizing subjects according to the presence/absence of residual symptoms, groups were compared along clinical variables and variables associated with residual symptoms were analyzed using multivariate analyses. Subjects with residual symptoms were less likely to report substance abuse (χ2(2) = 11.937, p ≤ 0.005) and lifetime psychotic symptoms (χ2(2) = 10.577, p = 0.005), and more likely to report higher illness episodes, longer duration of illness (t253 = 67.282, p ≤ 0.001; t253 = 10.755, p ≤ 0.001), and longer duration of current illness episode (t253 = 7.707, p ≤ 0.001) than those without residual symptoms. After multivariate analyses, a significant positive contribution to residual symptoms was given only by duration of current illness episode (β = 0.003; p ≤ 0.05), and lifetime psychotic symptoms (β = 1.094; p ≤ 0.005). Clinicians have to pay attention to minimize residual symptoms that may significantly impact on the course of BD and achievement of full remission between episodes.
Collapse
Affiliation(s)
- Gianluca Serafini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| | - Gustavo H Vazquez
- International Consortium for Bipolar and Psychotic Disorder Research, McLean Hospital, Belmont, MA, USA.,Department of Neuroscience, Palermo University, Buenos Aires, Argentina.,Department of Psychiatry, Queens University, Kingston, Ontario, Canada
| | - Xenia Gonda
- Department of Psychiatry and Psychotherapy, Kutvolgyi Clinical Center, Semmelweis University, Budapest, Hungary.,MTA-SE Neuropsychopharmacology Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary.,NAP-A-SE New Antidepressant Target Research Group, Semmelweis University, Budapest, Hungary
| | - Maurizio Pompili
- Department of Neurosciences, Suicide Prevention Center, Sant'Andrea Hospital, University of Rome, Rome, Italy
| | - Zoltan Rihmer
- Department of Psychiatry and Psychotherapy, Kutvolgyi Clinical Center, Semmelweis University, Budapest, Hungary
| | - Mario Amore
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), Section of Psychiatry, University of Genoa, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| |
Collapse
|
13
|
Martis LS, Brision C, Holmes MC, Wiborg O. Resilient and depressive-like rats show distinct cognitive impairments in the touchscreen paired-associates learning (PAL) task. Neurobiol Learn Mem 2018; 155:287-296. [PMID: 30138691 DOI: 10.1016/j.nlm.2018.08.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 08/06/2018] [Accepted: 08/18/2018] [Indexed: 01/17/2023]
Abstract
Depression-associated cognitive impairments persist after remission from affective symptoms of major depressive disorder (MDD), decreasing quality of life and increasing risk of relapse in patients. Conventional antidepressants are ineffective in restoring cognitive functions. Therefore, novel antidepressants with improved efficacy for ameliorating cognitive symptoms are required. For tailoring such antidepressants, translational animal models are in demand. The chronic mild stress (CMS) model is a well-validated preclinical model of depression and known for eliciting the MDD core symptom "anhedonia" in stress-susceptible rats. Thus, cognitive performance was assessed in rats susceptible (depressive-like) or resilient to CMS and in unchallenged controls. The rodent analogue of the human touchscreen Paired-Associates Learning (PAL) task was used for cognitive assessment. Both stress groups exhibited a lack of response inhibition compared to controls while only the depressive-like group was impaired in task acquisition. The results indicate that cognitive deficits specifically associate with the anhedonic-like state rather than being a general consequence of stress exposure. Hence, we propose that the application of a translational touchscreen task on the etiologically valid CMS model, displaying depression-associated cognitive impairments, provides a novel platform for pro-cognitive and clinically pertinent antidepressant drug screening.
Collapse
Affiliation(s)
- Lena-Sophie Martis
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark; Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Scotland, United Kingdom
| | - Claudia Brision
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Megan C Holmes
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Scotland, United Kingdom; Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Scotland, United Kingdom
| | - Ove Wiborg
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark; Department of Health Science and Technology, Aalborg University, Denmark.
| |
Collapse
|
14
|
Serafini G, Adavastro G, Canepa G, De Berardis D, Valchera A, Pompili M, Nasrallah H, Amore M. The Efficacy of Buprenorphine in Major Depression, Treatment-Resistant Depression and Suicidal Behavior: A Systematic Review. Int J Mol Sci 2018; 19:E2410. [PMID: 30111745 PMCID: PMC6121503 DOI: 10.3390/ijms19082410] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/31/2018] [Accepted: 08/08/2018] [Indexed: 01/08/2023] Open
Abstract
Although several pharmacological options to treat depression are currently available, approximately one third of patients who receive antidepressant medications do not respond adequately or achieve a complete remission. Thus, novel strategies are needed to successfully address those who did not respond, or partially respond, to available antidepressant pharmacotherapy. Research findings revealed that the opioid system is significantly involved in the regulation of mood and incentives salience and may be an appropriate target for novel therapeutic agents. The present study aimed to systematically review the current literature about the use of buprenorphine (BUP) for major depression, treatment-resistant depression (TRD), non-suicidal self-injury (NSSI) behavior, and suicidal behavior. We investigated Pubmed and Scopus databases using the following keywords: "buprenorphine AND depression", "buprenorphine AND treatment resistant depression", "buprenorphine AND suicid*", "buprenorphine AND refractory depression". Several evidence demonstrate that, at low doses, BUP is an efficacious, well-tolerated, and safe option in reducing depressive symptoms, serious suicidal ideation, and NSSI, even in patients with TRD. However, more studies are needed to evaluate the long-term effects, and relative efficacy of specific combinations (e.g., BUP + samidorphan (BUP/SAM), BUP + naloxone (BUP/NAL), BUP + naltrexone) over BUP monotherapy or adjunctive BUP treatment with standard antidepressants, as well as to obtain more uniform guidance about the optimal BUP dosing interval.
Collapse
Affiliation(s)
- Gianluca Serafini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, 16132 Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | - Giulia Adavastro
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, 16132 Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | - Giovanna Canepa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, 16132 Genoa, Italy.
| | - Domenico De Berardis
- Villa San Giuseppe Hospital, Hermanas Hospitalarias, Ascoli Piceno, Italy, Polyedra Research Group, 64100 Teramo, Italy.
| | - Alessandro Valchera
- NHS, Department of Mental Health, Psychiatric Service of Diagnosis and Treatment, Hospital "G. Mazzini", Asl 4, 64100 Teramo, Italy.
| | - Maurizio Pompili
- Department of Neurosciences, Mental Health and Sensory Organs, Suicide Prevention Center, Sant'Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy.
| | - Henry Nasrallah
- Department of Neurology & Psychiatry, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| | - Mario Amore
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, 16132 Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| |
Collapse
|
15
|
Gerhard DM, Duman RS. Rapid-Acting Antidepressants: Mechanistic Insights and Future Directions. Curr Behav Neurosci Rep 2018; 5:36-47. [PMID: 30034992 PMCID: PMC6051539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
PURPOSE OF REVIEW Ketamine produces rapid (within hours) antidepressant actions, even in patients considered treatment resistant, and even shows promise for suicidal ideation. Here, we review current research on the molecular and cellular mechanisms of ketamine and other novel rapid-acting antidepressants, and briefly explore gender differences in the pathophysiology and treatment of MDD. RECENT FINDINGS Ketamine, an NMDA receptor antagonist, increases BDNF release and synaptic connectivity, opposing the deficits caused by chronic stress and depression. Efforts are focused on the development of novel rapid agents that produce similar synaptic and rapid antidepressant actions, but without the side effects of ketamine. The impact of gender on the response to ketamine and other rapid-acting antidepressants is in early stages of investigation. SUMMARY The discovery that ketamine produces rapid therapeutic actions for depression and suicidal ideation represents a major breakthrough and much needed alternative to currently available medications. However, novel fast acting agents with fewer side effects are needed, as well as elucidation of the efficacy of these rapid-acting antidepressants for depression in women.
Collapse
Affiliation(s)
- Danielle M Gerhard
- Department of Psychiatry, Laboratory of Molecular Psychiatry, Yale University School of Medicine, New Haven, CT 06508, USA
| | - Ronald S Duman
- Department of Psychiatry, Laboratory of Molecular Psychiatry, Yale University School of Medicine, New Haven, CT 06508, USA
| |
Collapse
|
16
|
|
17
|
Scifo E, Pabba M, Kapadia F, Ma T, Lewis DA, Tseng GC, Sibille E. Sustained Molecular Pathology Across Episodes and Remission in Major Depressive Disorder. Biol Psychiatry 2018; 83:81-89. [PMID: 28935211 PMCID: PMC5705452 DOI: 10.1016/j.biopsych.2017.08.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 07/18/2017] [Accepted: 08/08/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is a debilitating mental illness and a major cause of lost productivity worldwide. MDD patients often suffer from lifelong recurring episodes of increasing severity, reduced therapeutic response, and shorter remission periods, suggesting the presence of a persistent and potentially progressive pathology. METHODS Subgenual anterior cingulate cortex postmortem samples from four MDD cohorts (single episode, n = 20; single episode in remission, n = 15; recurrent episode, n = 20; and recurrent episode in remission, n = 15), and one control cohort (n = 20) were analyzed by mass spectrometry-based proteomics (n = 3630 proteins) combined with statistical analyses. The data was investigated for trait and state progressive neuropathologies in MDD using both unbiased approaches and tests of a priori hypotheses. RESULTS The data provided weak evidence for proteomic differences as a function of state (depressed/remitted) or number of previous episodes. Instead it suggested the presence of persistent MDD effects, regardless of episodes or remitted state, namely on proteomic measures related to presynaptic neurotransmission, synaptic function, cytoskeletal rearrangements, energy metabolism, phospholipid biosynthesis/metabolism, and calcium ion homeostasis. Selected proteins (dihydropyrimidinase-related protein 1, synaptosomal-associated protein 29, glutamate decarboxylase 1, metabotropic glutamate receptor 1, and excitatory amino acid transporter 3) were validated by Western blot analysis. The findings were independent of technical, demographic (sex or age), or other clinical parameters (death by suicide and drug treatment). CONCLUSIONS Collectively, the results provide evidence for persistent MDD effects across current episodes or remission, in the absence of detectable progressive neuropathology.
Collapse
Affiliation(s)
- Enzo Scifo
- Campbell Family Mental Health Research Institute of CAMH, Department of Psychiatry, and of Pharmacology and Toxicology, University of Toronto, Toronto, M5T1R8, ON, Canada
| | - Mohan Pabba
- Campbell Family Mental Health Research Institute of CAMH, Department of Psychiatry, and of Pharmacology and Toxicology, University of Toronto, Toronto, M5T1R8, ON, Canada
| | - Fenika Kapadia
- Campbell Family Mental Health Research Institute of CAMH, Department of Psychiatry, and of Pharmacology and Toxicology, University of Toronto, Toronto, M5T1R8, ON, Canada
| | - Tianzhou Ma
- Department of Biostatistics, Graduate school of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - David A. Lewis
- Department of Psychiatry, 3811 O’Hara Street, BST W1643, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - George C Tseng
- Department of Biostatistics, Graduate school of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of the Centre for Addiction and Mental Health, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
18
|
Franklin TC, Wohleb ES, Zhang Y, Fogaça M, Hare B, Duman RS. Persistent Increase in Microglial RAGE Contributes to Chronic Stress-Induced Priming of Depressive-like Behavior. Biol Psychiatry 2018; 83:50-60. [PMID: 28882317 PMCID: PMC6369917 DOI: 10.1016/j.biopsych.2017.06.034] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 06/20/2017] [Accepted: 06/28/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Chronic stress-induced inflammatory responses occur in part via danger-associated molecular pattern (DAMP) molecules, such as high mobility group box 1 protein (HMGB1), but the receptor(s) underlying DAMP signaling have not been identified. METHODS Microglia morphology and DAMP signaling in enriched rat hippocampal microglia were examined during the development and expression of chronic unpredictable stress (CUS)-induced behavioral deficits, including long-term, persistent changes after CUS. RESULTS The results show that CUS promotes significant morphological changes and causes robust upregulation of HMGB1 messenger RNA in enriched hippocampal microglia, an effect that persists for up to 6 weeks after CUS exposure. This coincides with robust and persistent upregulation of receptor for advanced glycation end products (RAGE) messenger RNA, but not toll-like receptor 4 in hippocampal microglia. CUS also increased surface expression of RAGE protein on hippocampal microglia as determined by flow cytometry and returned to basal levels 5 weeks after CUS. Importantly, exposure to short-term stress was sufficient to increase RAGE surface expression as well as anhedonic behavior, reflecting a primed state that results from a persistent increase in RAGE messenger RNA expression. Further evidence for DAMP signaling in behavioral responses is provided by evidence that HMGB1 infusion into the hippocampus was sufficient to cause anhedonic behavior and by evidence that RAGE knockout mice were resilient to stress-induced anhedonia. CONCLUSIONS Together, the results provide evidence of persistent microglial HMGB1-RAGE expression that increases vulnerability to depressive-like behaviors long after chronic stress exposure.
Collapse
|
19
|
Fee C, Banasr M, Sibille E. Somatostatin-Positive Gamma-Aminobutyric Acid Interneuron Deficits in Depression: Cortical Microcircuit and Therapeutic Perspectives. Biol Psychiatry 2017; 82:549-559. [PMID: 28697889 PMCID: PMC5610074 DOI: 10.1016/j.biopsych.2017.05.024] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/11/2017] [Accepted: 05/30/2017] [Indexed: 12/17/2022]
Abstract
The functional integration of external and internal signals forms the basis of information processing and is essential for higher cognitive functions. This occurs in finely tuned cortical microcircuits whose functions are balanced at the cellular level by excitatory glutamatergic pyramidal neurons and inhibitory gamma-aminobutyric acidergic (GABAergic) interneurons. The balance of excitation and inhibition, from cellular processes to neural network activity, is characteristically disrupted in multiple neuropsychiatric disorders, including major depressive disorder (MDD), bipolar disorder, anxiety disorders, and schizophrenia. Specifically, nearly 3 decades of research demonstrate a role for reduced inhibitory GABA level and function across disorders. In MDD, recent evidence from human postmortem and animal studies suggests a selective vulnerability of GABAergic interneurons that coexpress the neuropeptide somatostatin (SST). Advances in cell type-specific molecular genetics have now helped to elucidate several important roles for SST interneurons in cortical processing (regulation of pyramidal cell excitatory input) and behavioral control (mood and cognition). Here, we review evidence for altered inhibitory function arising from GABAergic deficits across disorders and specifically in MDD. We then focus on properties of the cortical microcircuit, where SST-positive GABAergic interneuron deficits may disrupt functioning in several ways. Finally, we discuss the putative origins of SST cell deficits, as informed by recent research, and implications for therapeutic approaches. We conclude that deficits in SST interneurons represent a contributing cellular pathology and therefore a promising target for normalizing altered inhibitory function in MDD and other disorders with reduced SST cell and GABA functions.
Collapse
Affiliation(s)
- Corey Fee
- Campbell Family Mental Health Research Institute of Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Mounira Banasr
- Campbell Family Mental Health Research Institute of Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute of Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
20
|
Puralewski R, Vasilakis G, Seney ML. Sex-related factors influence expression of mood-related genes in the basolateral amygdala differentially depending on age and stress exposure. Biol Sex Differ 2016; 7:50. [PMID: 27660699 PMCID: PMC5027117 DOI: 10.1186/s13293-016-0106-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/09/2016] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Women are twice as likely to be diagnosed with major depressive disorder (MDD) compared to men, but the molecular mechanisms underlying this sex difference are unclear. Previous studies in the human postmortem brain suggest dysfunction in basolateral amygdala (BLA) inhibitory gamma-aminobutyric acid (GABA) signaling and brain-derived neurotrophic factor (BDNF) function, specifically in females with MDD. METHODS We investigated the effects of sex chromosome complement, developmental gonadal sex, and circulating testosterone on expression of 3 GABA-related and 2 BDNF-related genes in the BLA using three cohorts of four core genotypes (FCG) mice. Cohort 1 included gonadally intact pre-pubertal FCG mice; results were analyzed using two-way ANOVA (sex chromosome complement-by-gonadal sex). We examined the same genes under adult non-stressed (cohort 2) and chronically stressed conditions (cohort 3). The results for cohorts 2 and 3 were analyzed by three-way ANOVA (sex chromosome complement-by-gonadal sex-by-hormone). The use of heatmaps and Spearman correlation of BLA gene expression and anxiety-like behavior provides a global interpretation of gene expression patterns. RESULTS In weanlings, we found an effect of sex chromosome complement, with lower expression of GABA/BDNF-related genes in XY mice. Most of these effects did not persist into adulthood, although a number of interesting interactions between organizational and activational effects of hormones emerged. In our adult cohorts, we found that testosterone had different effects depending on stress conditions and/or gonadal sex. Notably, in our chronically stressed adults, we found that the BLA pattern of gene expression for the GABA-related gene, somatostatin (Sst), matched the anxiety-like behavior pattern (i.e., lower Sst and higher anxiety-like behavior in XY mice, while testosterone increased Sst and decreased anxiety-like behavior). Additionally, increased Sst gene expression was correlated with decreased anxiety-like behavior. CONCLUSIONS Sex chromosome complement is an important factor modulating expression of mood-related genes during pre-pubertal development. The observed sex differences under chronically stressed conditions suggest that different molecular profiles may characterize male and female MDD. Our findings here for Sst are especially interesting, and suggest an underlying XY vulnerability that is typically compensated for by circulating testosterone in "normal" males. Without testosterone, women may have lower SST expression in the amygdala, resulting in increased MDD vulnerability.
Collapse
Affiliation(s)
- Rachel Puralewski
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213 USA ; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Georgia Vasilakis
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213 USA ; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Marianne L Seney
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213 USA ; Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA 15213 USA
| |
Collapse
|
21
|
García-Fuster MJ, García-Sevilla JA. Effects of anti-depressant treatments on FADD and p-FADD protein in rat brain cortex: enhanced anti-apoptotic p-FADD/FADD ratio after chronic desipramine and fluoxetine administration. Psychopharmacology (Berl) 2016; 233:2955-71. [PMID: 27259485 DOI: 10.1007/s00213-016-4342-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/20/2016] [Indexed: 12/27/2022]
Abstract
RATIONALE Fas-associated death domain (FADD) is an adaptor of death receptors that can also induce anti-apoptotic actions through its phosphorylated form (p-FADD). Activation of monoamine receptors, indirect targets of classic anti-depressant drugs (ADs), reduced FADD and increased p-FADD and p-FADD/FADD ratio in brain. OBJECTIVES To ascertain whether ADs, which indirectly regulate monoamine receptors, modulate FADD protein forms to promote anti-apoptotic actions. METHODS The effects of selected norepinephrine transporter (NET), serotonin transporter (SERT), monoamine oxidase (MAO) inhibitors, atypical ADs, and electroconvulsive shock (ECS) or behavioral procedures (forced swim test, FST) on FADD forms and pro-survival FADD-like interleukin-1β-converting enzyme-inhibitory protein (FLIP-L) and phosphoprotein enriched in astrocytes of 15 kDa (p-PEA-15) contents were assessed in rat brain cortex by western blot analysis. RESULTS Acute NET (e.g., nisoxetine) but not SERT (e.g., fluoxetine) inhibitors decreased cortical FADD (up to 37 %) and increased p-FADD/FADD ratio (up to 1.9-fold). Nisoxetine effects were prevented by α2-antagonist RX-821002, suggesting the involvement of presynaptic α2-autoreceptors. Immobility time in the FST correlated with increases of pro-apoptotic FADD and decreases of anti-apoptotic p-FADD. The MAO-A/B inhibitor phenelzine decreased FADD (up to 33 %) and increased p-FADD (up to 65 %) and p-FADD/FADD (up to 2.4-fold). Other MAO inhibitors (clorgyline, Ro 41-1049, rasagiline), atypical ADs (ketamine and mirtazapine), or ECS did not modulate cortical FADD. Chronic (14 days) desipramine and fluoxetine, but not phenelzine, increased p-FADD (up to 59 %), p-FADD/FADD ratio (up to 1.8-fold), and pro-survival p-PEA-15 (up to 46 %) in rat brain cortex. CONCLUSIONS Multifunctional FADD protein, through an increased p-FADD/FADD ratio, could participate in the mechanisms of anti-apoptotic actions induced by ADs.
Collapse
Affiliation(s)
- M Julia García-Fuster
- Neurobiology of Drug Abuse Group, IUNICS/IdISPa, University of the Balearic Islands, Cra. Valldemossa km 7.5, E-07122, Palma de Mallorca, Spain. .,Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain.
| | - Jesús A García-Sevilla
- Laboratory of Neuropharmacology, IUNICS/IdISPa, University of the Balearic Islands, Palma de Mallorca, Spain.,Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| |
Collapse
|
22
|
Si T, Yu X. Current Problems in the Research and Development of more Effective Antidepressants. SHANGHAI ARCHIVES OF PSYCHIATRY 2016. [PMID: 28638186 PMCID: PMC5434302 DOI: 10.11919/j.issn.1002-0829.216017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This commentary was stimulated by discussions held at the First China Antidepressants Research and Development Summit held in Beijing in October 2015. Hosted by the Chinese Psychiatrist Psychopharmacology Commission and including leading clinicians, neuroscientists, and representatives of the pharmaceutical industry, the summit focused on the major problems that are limiting the development of more effective antidepressant medications. In the absence of clear biomarkers of depression, clinicians must base treatment decisions on clinical phenomenology; the lack of clear biological targets results in currently available antidepressants that take a long time to be effective, have low rates of full remission, and high rates of relapse. Basic research on depression by neuroscientists in China is internationally recognized, but the vast proportion of candidate chemical compounds Chinese researchers propose as potential treatments for depression fail when tested clinically. This high failure rate of proposed agents has rapidly increased the cost of bringing new drugs to market, so pharmaceutical firms prefer to ‘tweak’ currently approved medications rather than take the financial risk of supporting the development of novel antidepressants. Thus, the development of new, more effective treatments for depression is at a stalemate. Given the huge impact of depression on the economic development of China and other countries, it is essential to actively solicit the support of governments and communities in the efforts of clinicians, researchers, and the pharmaceutical industry to overcome this stalemate.
Collapse
Affiliation(s)
- Tianmei Si
- Peking University Institute of Mental Health (the Sixth Hospital), Beijing, China.,National Clinical Medical Research Center for Psychiatric Disorders and National Key Laboratory for Mental Health, Beijing, China
| | - Xin Yu
- Peking University Institute of Mental Health (the Sixth Hospital), Beijing, China.,National Clinical Medical Research Center for Psychiatric Disorders and National Key Laboratory for Mental Health, Beijing, China
| |
Collapse
|
23
|
Kim HK, Nunes PV, Oliveira KC, Young LT, Lafer B. Neuropathological relationship between major depression and dementia: A hypothetical model and review. Prog Neuropsychopharmacol Biol Psychiatry 2016; 67:51-7. [PMID: 26780170 DOI: 10.1016/j.pnpbp.2016.01.008] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/10/2016] [Accepted: 01/13/2016] [Indexed: 01/18/2023]
Abstract
Major depression (MDD) is a chronic psychiatric condition in which patients often show increasing cognitive impairment with recurring episodes. Neurodegeneration may play an important component in the pathogenesis of MDD associated with cognitive complaints. In agreement with this, patients with MDD show decreased brain volumes in areas implicated in emotional regulation and cognition, neuronal and glial cell death as well as activation of various pathways that can contribute to cell death. Therefore, the aim of this review is to provide an integrative overview of potential contributing factors to neurodegeneration in MDD. Studies have reported increased neuronal and glial cell death in the frontal cortex, amygdala, and hippocampus of patients with MDD. This may be due to decreased neurogenesis from lower levels of brain-derived neurotrophic factor (BDNF), excitotoxicity from increased glutamate signaling, and lower levels of gamma-aminobutyric acid (GABA) signaling. In addition, mitochondrial dysfunction and oxidative stress are found in similar brain areas where evidence of excitotoxicity has been reported. Also, levels of antioxidant enzymes were reported to be increased in patients with MDD. Inflammation may also be a contributing factor, as levels of inflammatory cytokines were reported to be increased in the prefrontal cortex of patients with MDD. While preliminary, studies have also reported neuropathological alterations in patients with MDD. Together, these studies suggest that lower BDNF levels, mitochondrial dysfunction, oxidative stress, inflammation and excitotoxicity may be contributing to neuronal and glial cell death in MDD, leading to decreased brain volume and cognitive dysfunction with multiple recurrent episodes. This highlights the need to identify specific pathways involved in neurodegeneration in MDD, which may elucidate targets that can be treated to ameliorate the effects of disease progression in this disorder.
Collapse
Affiliation(s)
- Helena Kyunghee Kim
- Departments of Psychiatry and Pharmacology, University of Toronto, RM4204, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.
| | - Paula Villela Nunes
- Bipolar Disorder Program (PROMAN), Department of Psychiatry, University of São Paulo Medical School, Rua Dr. Ovídio Pires de Campos, 785, São Paulo, 3671, Brazil.
| | - Katia C Oliveira
- Bipolar Disorder Program (PROMAN), Department of Psychiatry, University of São Paulo Medical School, Rua Dr. Ovídio Pires de Campos, 785, São Paulo, 3671, Brazil.
| | - L Trevor Young
- Departments of Psychiatry and Pharmacology, University of Toronto, RM4204, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.
| | - Beny Lafer
- Bipolar Disorder Program (PROMAN), Department of Psychiatry, University of São Paulo Medical School, Rua Dr. Ovídio Pires de Campos, 785, São Paulo, 3671, Brazil.
| |
Collapse
|
24
|
Mazereeuw G, Herrmann N, Andreazza AC, Khan MM, Lanctôt KL. A meta-analysis of lipid peroxidation markers in major depression. Neuropsychiatr Dis Treat 2015; 11:2479-91. [PMID: 26491326 PMCID: PMC4599178 DOI: 10.2147/ndt.s89922] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Major depressive disorder (MDD) may be associated with oxidative damage to lipids, which can potentially affect mood-regulating pathways. This meta-analysis summarizes current knowledge regarding lipid peroxidation markers in clinical samples of MDD and the effects of antidepressant pharmacotherapy on those markers. METHODS MEDLINE, EMBASE, CINAHL, PsycINFO, and Cochrane Collaboration were searched for original, peer-reviewed articles measuring markers of lipid peroxidation in patients with MDD and nondepressed healthy controls up to April 2015. Standardized mean differences (SMDs) were generated from random effects models summarizing mean (± standard deviations) concentrations of selected markers. RESULTS Lipid peroxidation was greater in MDD than in controls (studies =17, N=857 MDD/782 control, SMD =0.83 [0.56-1.09], z=6.11, P<0.01, I (2)=84.0%) and was correlated with greater depressive symptom severity (B=0.05, df=8, P<0.01). Antidepressant treatment was associated with a reduction in lipid peroxidation in MDD patients (studies=5, N=222, SMD=0.71 [0.40-0.97], P<0.01; I (2)=42.5%). LIMITATIONS Lipid peroxidation markers were sampled from peripheral blood, included studies comparing MDD to controls were all cross-sectional, and only five antidepressant treatment studies were eligible for inclusion. CONCLUSION Increased lipid peroxidation was associated with MDD and may be normalized by antidepressants. Continued investigation of lipid peroxidation in MDD is warranted.
Collapse
Affiliation(s)
- Graham Mazereeuw
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada ; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Nathan Herrmann
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada ; Department of Psychiatry, University of Toronto
| | - Ana C Andreazza
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada ; Department of Psychiatry, University of Toronto ; Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Maisha M Khan
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Krista L Lanctôt
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada ; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada ; Department of Psychiatry, University of Toronto
| |
Collapse
|
25
|
Lopizzo N, Bocchio Chiavetto L, Cattane N, Plazzotta G, Tarazi FI, Pariante CM, Riva MA, Cattaneo A. Gene-environment interaction in major depression: focus on experience-dependent biological systems. Front Psychiatry 2015; 6:68. [PMID: 26005424 PMCID: PMC4424810 DOI: 10.3389/fpsyt.2015.00068] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/21/2015] [Indexed: 12/27/2022] Open
Abstract
Major depressive disorder (MDD) is a multifactorial and polygenic disorder, where multiple and partially overlapping sets of susceptibility genes interact each other and with the environment, predisposing individuals to the development of the illness. Thus, MDD results from a complex interplay of vulnerability genes and environmental factors that act cumulatively throughout individual's lifetime. Among these environmental factors, stressful life experiences, especially those occurring early in life, have been suggested to exert a crucial impact on brain development, leading to permanent functional changes that may contribute to lifelong risk for mental health outcomes. In this review, we will discuss how genetic variants (polymorphisms, SNPs) within genes operating in neurobiological systems that mediate stress response and synaptic plasticity, can impact, by themselves, the vulnerability risk for MDD; we will also consider how this MDD risk can be further modulated when gene × environment interaction is taken into account. Finally, we will discuss the role of epigenetic mechanisms, and in particular of DNA methylation and miRNAs expression changes, in mediating the effect of the stress on the vulnerability risk to develop MDD. Taken together, we aim to underlie the role of genetic and epigenetic processes involved in stress- and neuroplasticity-related biological systems on the development of MDD after exposure to early life stress, thereby building the basis for future research and clinical interventions.
Collapse
Affiliation(s)
- Nicola Lopizzo
- IRCCS Fatebenefratelli San Giovanni di Dio , Brescia , Italy
| | - Luisella Bocchio Chiavetto
- IRCCS Fatebenefratelli San Giovanni di Dio , Brescia , Italy ; Faculty of Psychology, eCampus University , Novedrate, Como , Italy
| | - Nadia Cattane
- IRCCS Fatebenefratelli San Giovanni di Dio , Brescia , Italy
| | - Giona Plazzotta
- IRCCS Fatebenefratelli San Giovanni di Dio , Brescia , Italy
| | - Frank I Tarazi
- Department of Psychiatry and Neuroscience Program, McLean Hospital, Harvard Medical School , Belmont, MA , USA
| | - Carmine M Pariante
- Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, King's College London , London , UK
| | - Marco A Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan , Milan , Italy
| | - Annamaria Cattaneo
- IRCCS Fatebenefratelli San Giovanni di Dio , Brescia , Italy ; Stress, Psychiatry and Immunology Laboratory, Department of Psychological Medicine, Institute of Psychiatry, King's College London , London , UK
| |
Collapse
|
26
|
Saltiel PF, Silvershein DI. Major depressive disorder: mechanism-based prescribing for personalized medicine. Neuropsychiatr Dis Treat 2015; 11:875-88. [PMID: 25848287 PMCID: PMC4386790 DOI: 10.2147/ndt.s73261] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Individual patients with depression present with unique symptom clusters - before, during, and even after treatment. The prevalence of persistent, unresolved symptoms and their contribution to patient functioning and disease progression emphasize the importance of finding the right treatment choice at the onset and the utility of switching medications based on suboptimal responses. Our primary goal as clinicians is to improve patient function and quality of life. In fact, feelings of well-being and the return to premorbid levels of functioning are frequently rated by patients as being more important than symptom relief. However, functional improvements often lag behind resolution of mood, attributed in large part to persistent and functionally impairing symptoms - namely, fatigue, sleep/wake disturbance, and cognitive dysfunction. Thus, patient outcomes can be optimized by deconstructing each patient's depressive profile to its component symptoms and specifically targeting those domains that differentially limit patient function. This article will provide an evidence-based framework within which clinicians may tailor pharmacotherapy to patient symptomatology for improved treatment outcomes.
Collapse
Affiliation(s)
- Philip F Saltiel
- Department of Psychiatry, New York University School of Medicine/Langone Medical Center New York University Behavioral Health Programs, New York University Pearl Barlow Center for Memory Evaluation and Treatment, New York, NY, USA
| | - Daniel I Silvershein
- Department of Medicine, New York University School of Medicine/Langone Medical Center, New York, NY, USA
| |
Collapse
|
27
|
FADD adaptor and PEA-15/ERK1/2 partners in major depression and schizophrenia postmortem brains: basal contents and effects of psychotropic treatments. Neuroscience 2014; 277:541-51. [PMID: 25075716 DOI: 10.1016/j.neuroscience.2014.07.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 06/26/2014] [Accepted: 07/18/2014] [Indexed: 01/05/2023]
Abstract
Enhanced brain apoptosis (neurons and glia) may be involved in major depression (MD) and schizophrenia (SZ), mainly through the activation of the intrinsic (mitochondrial) apoptotic pathway. In the extrinsic death pathway, pro-apoptotic Fas-associated death domain (FADD) adaptor and its non-apoptotic p-Ser194 FADD form have critical roles interacting with other death regulators such as phosphoprotein enriched in astrocytes of 15 kDa (PEA-15) and extracellular signal-regulated kinase (ERK). The basal status of FADD (protein and messenger RNA (mRNA)) and the effects of psychotropic drugs (detected in blood/urine samples) were first assessed in postmortem prefrontal cortex of MD and SZ subjects (including a non-MD/SZ suicide group). In MD, p-FADD, but not total FADD (and mRNA), was increased (26%, n=24; all MD subjects) as well as p-FADD/FADD ratio (a pro-survival marker) in antidepressant-free MD subjects (50%, n=10). In contrast, cortical FADD (and mRNA), p-FADD, and p-FADD/FADD were not altered in SZ brains (n=21) regardless of antipsychotic medications (except enhanced mRNA in treated subjects). Similar negative results were quantified in the non-MD/SZ suicide group. In MD, the regulation of multifunctional PEA-15 (i.e., p-Ser116 PEA-15 blocks pro-apoptotic FADD and PEA-15 prevents pro-survival ERK action) and the modulation of p-ERK1/2 were also investigated. Cortical p-PEA-15 was not changed whereas PEA-15 was increased mainly in antidepressant-treated subjects (16-20%). Interestingly, cortical p-ERK1/2/ERK1/2 ratio was reduced (33%) in antidepressant-free when compared to antidepressant-treated MD subjects. The neurochemical adaptations of brain FADD (increased p-FADD and pro-survival p-FADD/FADD ratio), as well as its interaction with PEA-15, could play a major role to counteract the known activation of the mitochondrial apoptotic pathway in MD.
Collapse
|
28
|
Chang LC, Jamain S, Lin CW, Rujescu D, Tseng GC, Sibille E. A conserved BDNF, glutamate- and GABA-enriched gene module related to human depression identified by coexpression meta-analysis and DNA variant genome-wide association studies. PLoS One 2014; 9:e90980. [PMID: 24608543 PMCID: PMC3946570 DOI: 10.1371/journal.pone.0090980] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 01/31/2014] [Indexed: 11/29/2022] Open
Abstract
Large scale gene expression (transcriptome) analysis and genome-wide association studies (GWAS) for single nucleotide polymorphisms have generated a considerable amount of gene- and disease-related information, but heterogeneity and various sources of noise have limited the discovery of disease mechanisms. As systematic dataset integration is becoming essential, we developed methods and performed meta-clustering of gene coexpression links in 11 transcriptome studies from postmortem brains of human subjects with major depressive disorder (MDD) and non-psychiatric control subjects. We next sought enrichment in the top 50 meta-analyzed coexpression modules for genes otherwise identified by GWAS for various sets of disorders. One coexpression module of 88 genes was consistently and significantly associated with GWAS for MDD, other neuropsychiatric disorders and brain functions, and for medical illnesses with elevated clinical risk of depression, but not for other diseases. In support of the superior discriminative power of this novel approach, we observed no significant enrichment for GWAS-related genes in coexpression modules extracted from single studies or in meta-modules using gene expression data from non-psychiatric control subjects. Genes in the identified module encode proteins implicated in neuronal signaling and structure, including glutamate metabotropic receptors (GRM1, GRM7), GABA receptors (GABRA2, GABRA4), and neurotrophic and development-related proteins [BDNF, reelin (RELN), Ephrin receptors (EPHA3, EPHA5)]. These results are consistent with the current understanding of molecular mechanisms of MDD and provide a set of putative interacting molecular partners, potentially reflecting components of a functional module across cells and biological pathways that are synchronously recruited in MDD, other brain disorders and MDD-related illnesses. Collectively, this study demonstrates the importance of integrating transcriptome data, gene coexpression modules and GWAS results for providing novel and complementary approaches to investigate the molecular pathology of MDD and other complex brain disorders.
Collapse
MESH Headings
- Aged
- Brain/metabolism
- Brain/physiopathology
- Brain-Derived Neurotrophic Factor/genetics
- Brain-Derived Neurotrophic Factor/metabolism
- Case-Control Studies
- Cell Adhesion Molecules, Neuronal/genetics
- Cell Adhesion Molecules, Neuronal/metabolism
- Depressive Disorder, Major/genetics
- Depressive Disorder, Major/metabolism
- Depressive Disorder, Major/physiopathology
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/metabolism
- Female
- Gene Expression Profiling
- Gene Expression Regulation
- Gene Regulatory Networks
- Genetic Predisposition to Disease
- Genome, Human
- Genome-Wide Association Study
- Humans
- Male
- Metabolic Networks and Pathways/genetics
- Middle Aged
- Multigene Family
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Receptors, Eph Family/genetics
- Receptors, Eph Family/metabolism
- Receptors, GABA/genetics
- Receptors, GABA/metabolism
- Receptors, Metabotropic Glutamate/genetics
- Receptors, Metabotropic Glutamate/metabolism
- Reelin Protein
- Serine Endopeptidases/genetics
- Serine Endopeptidases/metabolism
- Transcriptome
Collapse
Affiliation(s)
- Lun-Ching Chang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stephane Jamain
- Inserm U955, Psychiatrie Génétique, Créteil, France
- Université Paris Est, Créteil, France
- Fondation FondaMental, Créteil, France
| | - Chien-Wei Lin
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Dan Rujescu
- Department of Psychiatry, University of Halle, Halle, Germany
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Etienne Sibille
- Department of Psychiatry, Center For Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
29
|
Gaiteri C, Ding Y, French B, Tseng GC, Sibille E. Beyond modules and hubs: the potential of gene coexpression networks for investigating molecular mechanisms of complex brain disorders. GENES, BRAIN, AND BEHAVIOR 2014; 13:13-24. [PMID: 24320616 PMCID: PMC3896950 DOI: 10.1111/gbb.12106] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 09/25/2013] [Accepted: 11/10/2013] [Indexed: 12/12/2022]
Abstract
In a research environment dominated by reductionist approaches to brain disease mechanisms, gene network analysis provides a complementary framework in which to tackle the complex dysregulations that occur in neuropsychiatric and other neurological disorders. Gene-gene expression correlations are a common source of molecular networks because they can be extracted from high-dimensional disease data and encapsulate the activity of multiple regulatory systems. However, the analysis of gene coexpression patterns is often treated as a mechanistic black box, in which looming 'hub genes' direct cellular networks, and where other features are obscured. By examining the biophysical bases of coexpression and gene regulatory changes that occur in disease, recent studies suggest it is possible to use coexpression networks as a multi-omic screening procedure to generate novel hypotheses for disease mechanisms. Because technical processing steps can affect the outcome and interpretation of coexpression networks, we examine the assumptions and alternatives to common patterns of coexpression analysis and discuss additional topics such as acceptable datasets for coexpression analysis, the robust identification of modules, disease-related prioritization of genes and molecular systems and network meta-analysis. To accelerate coexpression research beyond modules and hubs, we highlight some emerging directions for coexpression network research that are especially relevant to complex brain disease, including the centrality-lethality relationship, integration with machine learning approaches and network pharmacology.
Collapse
Affiliation(s)
- Chris Gaiteri
- . Modeling, Analysis and Theory Group, Allen Institute for Brain Science, Seattle WA, USA
| | - Ying Ding
- . Carnegie Mellon-University of Pittsburgh PhD Program in Computational Biology, Pittsburgh, PA, USA
| | - Beverly French
- . Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - George C. Tseng
- . Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Etienne Sibille
- . Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
30
|
Patel S. Role of Proteomics in Biomarker Discovery. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2014; 94:39-75. [DOI: 10.1016/b978-0-12-800168-4.00003-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|