51
|
Xu W, Liu X, Leng F, Li W. Blood-based multi-tissue gene expression inference with Bayesian ridge regression. Bioinformatics 2020; 36:3788-3794. [PMID: 32277818 DOI: 10.1093/bioinformatics/btaa239] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 03/15/2020] [Accepted: 04/06/2020] [Indexed: 12/23/2022] Open
Abstract
MOTIVATION Gene expression profiling is widely used in basic and cancer research but still not feasible in many clinical applications because tissues, such as brain samples, are difficult and not ethnical to collect. Gene expression in uncollected tissues can be computationally inferred using genotype and expression quantitative trait loci. No methods can infer unmeasured gene expression of multiple tissues with single tissue gene expression profile as input. RESULTS Here, we present a Bayesian ridge regression-based method (B-GEX) to infer gene expression profiles of multiple tissues from blood gene expression profile. For each gene in a tissue, a low-dimensional feature vector was extracted from whole blood gene expression profile by feature selection. We used GTEx RNAseq data of 16 tissues to train inference models to capture the cross-tissue expression correlations between each target gene in a tissue and its preselected feature genes in peripheral blood. We compared B-GEX with least square regression, LASSO regression and ridge regression. B-GEX outperforms the other three models in most tissues in terms of mean absolute error, Pearson correlation coefficient and root-mean-squared error. Moreover, B-GEX infers expression level of tissue-specific genes as well as those of non-tissue-specific genes in all tissues. Unlike previous methods, which require genomic features or gene expression profiles of multiple tissues, our model only requires whole blood expression profile as input. B-GEX helps gain insights into gene expressions of uncollected tissues from more accessible data of blood. AVAILABILITY AND IMPLEMENTATION B-GEX is available at https://github.com/xuwenjian85/B-GEX. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Wenjian Xu
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Genetics and Birth Defects Control Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Xuanshi Liu
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Genetics and Birth Defects Control Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Fei Leng
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Genetics and Birth Defects Control Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Wei Li
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Genetics and Birth Defects Control Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| |
Collapse
|
52
|
Manosso LM, Camargo A, Dafre AL, Rodrigues ALS. Vitamin E for the management of major depressive disorder: possible role of the anti-inflammatory and antioxidant systems. Nutr Neurosci 2020; 25:1310-1324. [PMID: 33314993 DOI: 10.1080/1028415x.2020.1853417] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Vitamin E has various functions in humans, including antioxidant, anti-inflammatory, anti-cancer, and anti-atherogenic actions, as well as direct effects on enzymatic activities and modulation of gene transcription. In addition to these functions, vitamin E is also important for the central nervous system, and its role in the prevention and/or treatment of some neurological diseases has been suggested. In particular, the role of vitamin E in the modulation of major depressive disorder (MDD) is an issue that has emerged in recent studies. Many factors have been implicated in the pathophysiology of this disorder, including inflammation, oxidative, and nitrosative stress. METHODS This narrative review discusses the involvement of inflammation, oxidative, and nitrosative stress in the pathophysiology of MDD and presents clinical and preclinical studies that correlate vitamin E with this psychiatric disorder. RESULTS We gathered evidence from clinical studies that demonstrated the relationship between low vitamin E status and MDD symptoms. Vitamin E has been reported to exert a beneficial influence on the oxidative and inflammatory status of individuals, factors that may account for the attenuation of depressive symptoms. Preclinical studies have reinforced the antidepressant-like response of vitamin E, and the mechanisms underlying its effect seem to be related to the modulation of oxidative stress and neuroinflammation. CONCLUSION We suggest that vitamin E has potential to be used as an adjuvant for the management of MDD, but more studies are clearly needed to ascertain the efficacy of vitamin E for alleviating depressive symptoms.
Collapse
Affiliation(s)
- Luana M Manosso
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Anderson Camargo
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Alcir L Dafre
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Ana Lúcia S Rodrigues
- Department of Biochemistry, Center of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| |
Collapse
|
53
|
Dmitrzak-Weglarz M, Szczepankiewicz A, Kapelski, Chaberska J, Kwiatkowska K, Duda J, Dziuda S, Skibinska M, Reszka E, Pawlak J. Transcripts of orphan nuclear receptor (NR4A1) & potassium channel (KCNK17) genes as new potential biomarkers for depression. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022] Open
|
54
|
Athira KV, Bandopadhyay S, Samudrala PK, Naidu VGM, Lahkar M, Chakravarty S. An Overview of the Heterogeneity of Major Depressive Disorder: Current Knowledge and Future Prospective. Curr Neuropharmacol 2020; 18:168-187. [PMID: 31573890 PMCID: PMC7327947 DOI: 10.2174/1570159x17666191001142934] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 08/05/2019] [Accepted: 09/27/2019] [Indexed: 02/08/2023] Open
Abstract
Major depressive disorder (MDD) is estimated to impose maximum debilitating effects on the society by 2030, with its critical effects on health, functioning, quality of life and concomitant high levels of morbidity and mortality. Yet, the disease is inadequately understood, diagnosed and treated. Moreover, with the recent drastic rise in the pace of life, stress has materialized as one of the most potent environmental factors for depression. In this scenario, it is important to understand the modern pathogenetic hypotheses and mechanisms, and possibly try to shift from the traditional approaches in depression therapy. These include the elaboration of pathophysiological changes in heterogeneous systems such as genetic, epigenetic, serotonergic, noradrenergic, gamma-aminobutyric acid, glutamatergic and endocannabinoid systems, neurotrophic factors, HPA axis, immune system as well as cellular stress mechanisms. These components interact with each other in a complex matrix and further elucidation of their mechanism and cascade pathways are needed. This might aid in the identification of MDD subtypes as well as the development of sophisticated biomarkers. Further, characterization might also aid in developing multitargeted therapies that hold much promise as compared to the conventional monoamine based treatment. New candidate pharmacons, refined psychotherapeutic modalities, advanced neuro-surgical and imaging techniques as well as the implementation of pharmacokinetic, pharmacogenetic prescribing guidelines constitute the emerging expanses of MDD treatment.
Collapse
Affiliation(s)
- Kaipuzha Venu Athira
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, 781125, Assam, India.,Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India.,Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682 041, Kerala, India
| | - Sikta Bandopadhyay
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| | - Pavan Kumar Samudrala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, 781125, Assam, India
| | - V G M Naidu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, 781125, Assam, India
| | - Mangala Lahkar
- Department of Pharmacology, Gauhati Medical College, Guwahati, 781032, Assam, India
| | - Sumana Chakravarty
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, India
| |
Collapse
|
55
|
Statins in the treatment of depression: Hype or hope? Pharmacol Ther 2020; 215:107625. [DOI: 10.1016/j.pharmthera.2020.107625] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/29/2020] [Indexed: 12/19/2022]
|
56
|
Wittenberg GM, Greene J, Vértes PE, Drevets WC, Bullmore ET. Major Depressive Disorder Is Associated With Differential Expression of Innate Immune and Neutrophil-Related Gene Networks in Peripheral Blood: A Quantitative Review of Whole-Genome Transcriptional Data From Case-Control Studies. Biol Psychiatry 2020; 88:625-637. [PMID: 32653108 DOI: 10.1016/j.biopsych.2020.05.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/11/2020] [Accepted: 05/03/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Whole-genome transcription has been measured in peripheral blood samples as a candidate biomarker of inflammation associated with major depressive disorder. METHODS We searched for all case-control studies on major depressive disorder that reported microarray or RNA sequencing measurements on whole blood or peripheral blood mononuclear cells. Primary datasets were reanalyzed, when openly accessible, to estimate case-control differences and to evaluate the functional roles of differentially expressed gene lists by technically harmonized methods. RESULTS We found 10 eligible studies (N = 1754 depressed cases and N = 1145 healthy controls). Fifty-two genes were called significant by 2 of the primary studies (published overlap list). After harmonization of analysis across 8 accessible datasets (n = 1706 cases, n = 1098 controls), 272 genes were coincidentally listed in the top 3% most differentially expressed genes in 2 or more studies of whole blood or peripheral blood mononuclear cells with concordant direction of effect (harmonized overlap list). By meta-analysis of standardized mean difference across 4 studies of whole-blood samples (n = 1567 cases, n = 954 controls), 343 genes were found with false discovery rate <5% (standardized mean difference meta-analysis list). These 3 lists intersected significantly. Genes abnormally expressed in major depressive disorder were enriched for innate immune-related functions, coded for nonrandom protein-protein interaction networks, and coexpressed in the normative transcriptome module specialized for innate immune and neutrophil functions. CONCLUSIONS Quantitative review of existing case-control data provided robust evidence for abnormal expression of gene networks important for the regulation and implementation of innate immune response. Further development of white blood cell transcriptional biomarkers for inflamed depression seems warranted.
Collapse
Affiliation(s)
- Gayle M Wittenberg
- Neuroscience, Janssen Research & Development, LLC, Titusville, New Jersey
| | - Jon Greene
- Bioinformatics, Rancho BioSciences, LLC, San Diego, California
| | - Petra E Vértes
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom; Alan Turing Institute, London, United Kingdom
| | - Wayne C Drevets
- Neuroscience, Janssen Research & Development, LLC, San Diego, California
| | - Edward T Bullmore
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom; Cambridgeshire and Peterborough National Health Service Foundation Trust, Cambridge, United Kingdom.
| |
Collapse
|
57
|
Kokkosis AG, Tsirka SE. Neuroimmune Mechanisms and Sex/Gender-Dependent Effects in the Pathophysiology of Mental Disorders. J Pharmacol Exp Ther 2020; 375:175-192. [PMID: 32661057 PMCID: PMC7569311 DOI: 10.1124/jpet.120.266163] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/09/2020] [Indexed: 12/12/2022] Open
Abstract
Innate and adaptive immune mechanisms have emerged as critical regulators of CNS homeostasis and mental health. A plethora of immunologic factors have been reported to interact with emotion- and behavior-related neuronal circuits, modulating susceptibility and resilience to mental disorders. However, it remains unclear whether immune dysregulation is a cardinal causal factor or an outcome of the pathologies associated with mental disorders. Emerging variations in immune regulatory pathways based on sex differences provide an additional framework for discussion in these psychiatric disorders. In this review, we present the current literature pertaining to the effects that disrupted immune pathways have in mental disorder pathophysiology, including immune dysregulation in CNS and periphery, microglial activation, and disturbances of the blood-brain barrier. In addition, we present the suggested origins of such immune dysregulation and discuss the gender and sex influence of the neuroimmune substrates that contribute to mental disorders. The findings challenge the conventional view of these disorders and open the window to a diverse spectrum of innovative therapeutic targets that focus on the immune-specific pathophenotypes in neuronal circuits and behavior. SIGNIFICANCE STATEMENT: The involvement of gender-dependent inflammatory mechanisms on the development of mental pathologies is gaining momentum. This review addresses these novel factors and presents the accumulating evidence introducing microglia and proinflammatory elements as critical components and potential targets for the treatment of mental disorders.
Collapse
Affiliation(s)
- Alexandros G Kokkosis
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| | - Stella E Tsirka
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York
| |
Collapse
|
58
|
Dattilo V, Amato R, Perrotti N, Gennarelli M. The Emerging Role of SGK1 (Serum- and Glucocorticoid-Regulated Kinase 1) in Major Depressive Disorder: Hypothesis and Mechanisms. Front Genet 2020; 11:826. [PMID: 32849818 PMCID: PMC7419621 DOI: 10.3389/fgene.2020.00826] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/09/2020] [Indexed: 12/28/2022] Open
Abstract
Major depressive disorder (MDD) is a heterogeneous psychiatric disease characterized by persistent low mood, diminished interests, and impaired cognitive and social functions. The multifactorial etiology of MDD is still largely unknown because of the complex genetic and environmental interactions involved. Therefore, no established mechanism can explain all the aspects of the disease. In this light, an extensive research about the pathophysiology of MDD has been carried out. Several pathogenic hypotheses, such as monoamines deficiency and neurobiological alterations in the stress-responsive system, including the hypothalamic-pituitary-adrenal (HPA) axis and the immune system, have been proposed for MDD. Over time, remarkable studies, mainly on preclinical rodent models, linked the serum- and glucocorticoid-regulated kinase 1 (SGK1) to the main features of MDD. SGK1 is a serine/threonine kinase belonging to the AGK Kinase family. SGK1 is ubiquitously expressed, which plays a pivotal role in the hormonal regulation of several ion channels, carriers, pumps, and transcription factors or regulators. SGK1 expression is modulated by cell stress and hormones, including gluco- and mineralocorticoids. Compelling evidence suggests that increased SGK1 expression or function is related to the pathogenic stress hypothesis of major depression. Therefore, the first part of the present review highlights the putative role of SGK1 as a critical mediator in the dysregulation of the HPA axis, observed under chronic stress conditions, and its controversial role in the neuroinflammation as well. The second part depicts the negative regulation exerted by SGK1 in the expression of both the brain-derived neurotrophic factor (BDNF) and the vascular endothelial growth factor (VEGF), resulting in an anti-neurogenic activity. Finally, the review focuses on the antidepressant-like effects of anti-oxidative nutraceuticals in several preclinical model of depression, resulting from the restoration of the physiological expression and/or activity of SGK1, which leads to an increase in neurogenesis. In summary, the purpose of this review is a systematic analysis of literature depicting SGK1 as molecular junction of the complex mechanisms underlying the MDD in an effort to suggest the kinase as a potential biomarker and strategic target in modern molecular antidepressant therapy.
Collapse
Affiliation(s)
- Vincenzo Dattilo
- Genetic Unit, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Rosario Amato
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Medical Genetics Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Nicola Perrotti
- Department of Health Sciences, Magna Graecia University of Catanzaro, Catanzaro, Italy.,Medical Genetics Unit, Mater Domini University Hospital, Catanzaro, Italy
| | - Massimo Gennarelli
- Genetic Unit, IRCCS Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
59
|
Tubbs JD, Ding J, Baum L, Sham PC. Immune dysregulation in depression: Evidence from genome-wide association. Brain Behav Immun Health 2020; 7:100108. [PMID: 34589869 PMCID: PMC8474691 DOI: 10.1016/j.bbih.2020.100108] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/12/2020] [Indexed: 12/15/2022] Open
Abstract
A strong body of evidence supports a role for immune dysregulation across many psychiatric disorders including depression, the leading cause of global disability. Recent progress in the search for genetic variants associated with depression provides the opportunity to strengthen our current understanding of etiological factors contributing to depression and generate novel hypotheses. Here, we provide an overview of the literature demonstrating a role for immune dysregulation in depression, followed by a detailed discussion of the immune-related genes identified by the most recent genome-wide meta-analysis of depression. These genes represent strong evidence-based targets for future basic and translational research which aims to understand the role of the immune system in depression pathology and identify novel points for therapeutic intervention.
Collapse
Affiliation(s)
- Justin D. Tubbs
- Department of Psychiatry, The University of Hong Kong, Hong Kong
| | - Jiahong Ding
- Department of Psychiatry, The University of Hong Kong, Hong Kong
| | - Larry Baum
- Department of Psychiatry, The University of Hong Kong, Hong Kong
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong
| | - Pak C. Sham
- Department of Psychiatry, The University of Hong Kong, Hong Kong
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong
- Centre for PanorOmic Sciences, The University of Hong Kong, Hong Kong
| |
Collapse
|
60
|
Wang D, Tang W, Zhao J, Fan W, Zhang Y, Zhang C. A Comprehensive Analysis of the Effect of SIRT1 Variation on the Risk of Schizophrenia and Depressive Symptoms. Front Genet 2020; 11:832. [PMID: 32849821 PMCID: PMC7413929 DOI: 10.3389/fgene.2020.00832] [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: 03/31/2020] [Accepted: 07/09/2020] [Indexed: 12/22/2022] Open
Abstract
Depressive symptoms could be considered a mutual manifestation of major depressive disorder and schizophrenia. Rs3758391 is a functional locus of Sirtuin (SIRT1) involving depression etiology. In this study, we hypothesized that the SIRT1 SNP rs3758391 might be a hazard for schizophrenia pathogenesis, especially related to the appearance of depressive symptoms. We recruited 723 healthy controls and 715 schizophrenia patients, the occurrence of psychotic and depressive symptoms was evaluated by Calgary Depression Scale (CDSS) and PANSS. Meanwhile, qt-PCR was used to detect the mRNA levels of SIRT1 in peripheral blood of 197 olanzapine monotherapy schizophrenia patients. 45.6% of schizophrenia patients had depressive symptoms. In the patient group, mRNA levels of patients with depressive symptoms were significantly lower than those without depressive symptoms (P < 0.01). CDSS scores of schizophrenia patients with different rs3758391 genotypes were significantly different (P < 0.01). Post hoc comparisons indicated that the CDSS scores of rs3758391 C/C and C/T carriers were higher than those of T/T carriers (Ps < 0.01). In the occipital cortex, our eQTL analysis showed that there was a clear correlation between rs3758391 and the SIRT1 mRNA levels. Our preliminary findings provide suggestive evidence that SIRT1 makes schizophrenia patients more prone to depressive symptoms. This SNP might be a biomarker of depression in schizophrenia.
Collapse
Affiliation(s)
- Dandan Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Tang
- Department of Psychiatry, The Affiliated Kangning Hospital of Wenzhou Medical University, Wenzhou, China
| | - Junxiong Zhao
- Department of Psychiatry, Jinhua Second Hospital, Jinhua, China
| | - Weixing Fan
- Department of Psychiatry, Jinhua Second Hospital, Jinhua, China
| | - Yi Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
61
|
Cattaneo A, Ferrari C, Turner L, Mariani N, Enache D, Hastings C, Kose M, Lombardo G, McLaughlin AP, Nettis MA, Nikkheslat N, Sforzini L, Worrell C, Zajkowska Z, Cattane N, Lopizzo N, Mazzelli M, Pointon L, Cowen PJ, Cavanagh J, Harrison NA, de Boer P, Jones D, Drevets WC, Mondelli V, Bullmore ET, Pariante CM. Whole-blood expression of inflammasome- and glucocorticoid-related mRNAs correctly separates treatment-resistant depressed patients from drug-free and responsive patients in the BIODEP study. Transl Psychiatry 2020; 10:232. [PMID: 32699209 PMCID: PMC7376244 DOI: 10.1038/s41398-020-00874-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
The mRNA expression signatures associated with the 'pro-inflammatory' phenotype of depression, and the differential signatures associated with depression subtypes and the effects of antidepressants, are still unknown. We examined 130 depressed patients (58 treatment-resistant, 36 antidepressant-responsive and 36 currently untreated) and 40 healthy controls from the BIODEP study, and used whole-blood mRNA qPCR to measure the expression of 16 candidate mRNAs, some never measured before: interleukin (IL)-1-beta, IL-6, TNF-alpha, macrophage inhibiting factor (MIF), glucocorticoid receptor (GR), SGK1, FKBP5, the purinergic receptor P2RX7, CCL2, CXCL12, c-reactive protein (CRP), alpha-2-macroglobulin (A2M), acquaporin-4 (AQP4), ISG15, STAT1 and USP-18. All genes but AQP4, ISG15 and USP-18 were differentially regulated. Treatment-resistant and drug-free depressed patients had both increased inflammasome activation (higher P2RX7 and proinflammatory cytokines/chemokines mRNAs expression) and glucocorticoid resistance (lower GR and higher FKBP5 mRNAs expression), while responsive patients had an intermediate phenotype with, additionally, lower CXCL12. Most interestingly, using binomial logistics models we found that a signature of six mRNAs (P2RX7, IL-1-beta, IL-6, TNF-alpha, CXCL12 and GR) distinguished treatment-resistant from responsive patients, even after adjusting for other variables that were different between groups, such as a trait- and state-anxiety, history of childhood maltreatment and serum CRP. Future studies should replicate these findings in larger, longitudinal cohorts, and test whether this mRNA signature can identify patients that are more likely to respond to adjuvant strategies for treatment-resistant depression, including combinations with anti-inflammatory medications.
Collapse
Affiliation(s)
- Annamaria Cattaneo
- Biological Psychiatric Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
| | - Clarissa Ferrari
- Statistical Service, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
| | - Lorinda Turner
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Nicole Mariani
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK
| | - Daniela Enache
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK
| | - Caitlin Hastings
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK
| | - Melisa Kose
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK
| | - Giulia Lombardo
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK
| | - Anna P McLaughlin
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK
| | - Maria A Nettis
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK
| | - Naghmeh Nikkheslat
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK
| | - Luca Sforzini
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK
| | - Courtney Worrell
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK
| | - Zuzanna Zajkowska
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK
| | - Nadia Cattane
- Biological Psychiatric Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
| | - Nicola Lopizzo
- Biological Psychiatric Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
| | - Monica Mazzelli
- Biological Psychiatric Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
| | - Linda Pointon
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Philip J Cowen
- University of Oxford Department of Psychiatry, Warneford Hospital, Oxford, OX3 7JX, UK
| | - Jonathan Cavanagh
- Centre for Immunobiology, University of Glasgow and Sackler Institute of Psychobiological Research, Queen Elizabeth University Hospital, Glasgow, G51 4TF, UK
| | - Neil A Harrison
- School of Medicine, School of Psychology, Cardiff University Brain Research Imaging Centre, Maindy Road, Cardiff, CF24 4HQ, UK
| | - Peter de Boer
- Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV, 2340, Beerse, Belgium
| | - Declan Jones
- Neuroscience External Innovation, Janssen Pharmaceuticals, J&J Innovation Centre, London, W1G 0BG, UK
| | - Wayne C Drevets
- Janssen Research & Development, Neuroscience Therapeutic Area, 3210 Merryfield Row, San Diego, CA, 92121, USA
| | - Valeria Mondelli
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK
| | - Edward T Bullmore
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Carmine M Pariante
- Stress, Psychiatry and Immunology Laboratory & Perinatal Psychiatry, King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, King's College London, SE5 9RT, London, UK.
| |
Collapse
|
62
|
The Bidirectional Relationship of Depression and Inflammation: Double Trouble. Neuron 2020; 107:234-256. [PMID: 32553197 DOI: 10.1016/j.neuron.2020.06.002] [Citation(s) in RCA: 1188] [Impact Index Per Article: 237.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/21/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022]
Abstract
Depression represents the number one cause of disability worldwide and is often fatal. Inflammatory processes have been implicated in the pathophysiology of depression. It is now well established that dysregulation of both the innate and adaptive immune systems occur in depressed patients and hinder favorable prognosis, including antidepressant responses. In this review, we describe how the immune system regulates mood and the potential causes of the dysregulated inflammatory responses in depressed patients. However, the proportion of never-treated major depressive disorder (MDD) patients who exhibit inflammation remains to be clarified, as the heterogeneity in inflammation findings may stem in part from examining MDD patients with varied interventions. Inflammation is likely a critical disease modifier, promoting susceptibility to depression. Controlling inflammation might provide an overall therapeutic benefit, regardless of whether it is secondary to early life trauma, a more acute stress response, microbiome alterations, a genetic diathesis, or a combination of these and other factors.
Collapse
|
63
|
Stonawski V, Roetner J, Goecke TW, Fasching PA, Beckmann MW, Kornhuber J, Kratz O, Moll GH, Eichler A, Heinrich H, Frey S. Genome-Wide DNA Methylation Patterns in Children Exposed to Nonpharmacologically Treated Prenatal Depressive Symptoms: Results From 2 Independent Cohorts. Epigenet Insights 2020; 13:2516865720932146. [PMID: 32596638 PMCID: PMC7298426 DOI: 10.1177/2516865720932146] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/01/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Maternal depressive symptoms are a common phenomenon during pregnancy and are related to negative outcomes for child development and health. Modifications in child DNA methylation are discussed as an underlying mechanism for the association between prenatal depressive symptoms and alterations in child outcomes. However, formerly reported genome-wide associations have yet to be replicated. METHODS In an epigenome-wide association study (EWAS), alterations of DNA methylation related to maternal prenatal depressive symptoms were investigated in buccal cell samples from 174 children (n = 52 exposed to prenatal depressive symptoms; 6-9 years old) of the German longitudinal study FRAMES-FRANCES. Whole blood samples from the independent, age-comparable ARIES subsample of the ARIES/ALSPAC study (n = 641; n = 159 exposed to prenatal depressive symptoms; 7-8 years old) were examined as a confirmation sample. Depressive symptoms were assessed with the Edinburgh Postnatal Depression Scale. DNA methylation was analyzed with the Infinium Human Methylation 450k BeadChip. Modifications in single CpGs, regions, and biological pathways were investigated. Results were adjusted for age and birth outcomes as well as postnatal and current maternal depressive symptoms. Analyses were performed for the whole sample as well as separated for sex. RESULTS The EWAS yielded no differentially methylated CpG or region as well as no accordance between samples withstanding correction for multiple testing. In pathway analyses, no overlapping functional domain was found to be enriched for either sample. A comparison of current and former findings suggests some overlapping methylation modifications from infancy to childhood. Results suggest that there might be sex-specific differential methylation, which should be further investigated in additional studies. CONCLUSIONS The current, mainly nonsignificant, results challenge the assumption of consistent modifications of DNA methylation in children exposed to prenatal depressive symptoms. Despite the relatively small sample size used in this study, this lack of significant results may reflect diverse issues of environmental epigenetic studies, which need to be addressed in future research.
Collapse
Affiliation(s)
- Valeska Stonawski
- Department of Child and Adolescent
Mental Health, University Hospital Erlangen, Friedrich-Alexander University
Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jakob Roetner
- Department of Child and Adolescent
Mental Health, University Hospital Erlangen, Friedrich-Alexander University
Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Tamme W Goecke
- Department of Gynecology and Obstetrics,
University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg
(FAU), Erlangen, Germany
- Department of Obstetrics and
Gynaecology, RoMed Hospital Rosenheim, Rosenheim, Germany
| | - Peter A Fasching
- Department of Gynecology and Obstetrics,
University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg
(FAU), Erlangen, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics,
University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg
(FAU), Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and
Psychotherapy, University Hospital Erlangen, Friedrich-Alexander University
Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Oliver Kratz
- Department of Child and Adolescent
Mental Health, University Hospital Erlangen, Friedrich-Alexander University
Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Gunther H Moll
- Department of Child and Adolescent
Mental Health, University Hospital Erlangen, Friedrich-Alexander University
Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anna Eichler
- Department of Child and Adolescent
Mental Health, University Hospital Erlangen, Friedrich-Alexander University
Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | - Stefan Frey
- Department of Child and Adolescent
Mental Health, University Hospital Erlangen, Friedrich-Alexander University
Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
64
|
Systemic immunization with altered myelin basic protein peptide produces sustained antidepressant-like effects. Mol Psychiatry 2020; 25:1260-1274. [PMID: 31375779 DOI: 10.1038/s41380-019-0470-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 06/17/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022]
Abstract
Immune dysregulation, specifically of inflammatory processes, has been linked to behavioral symptoms of depression in both human and rodent studies. Here, we evaluated the antidepressant effects of immunization with altered peptide ligands of myelin basic protein (MBP)-MBP87-99[A91, A96], MBP87-99[A91], and MBP87-99[R91, A96]-in different models of depression and examined the mechanism by which these peptides protect against stress-induced depression. We found that a single dose of subcutaneously administered MBP87-99[A91, A96] produced antidepressant-like effects by decreasing immobility in the forced swim test and by reducing the escape latency and escape failures in the learned helplessness paradigm. Moreover, immunization with MBP87-99[A91, A96] prevented and reversed depressive-like and anxiety-like behaviors that were induced by chronic unpredictable stress (CUS). However, MBP87-99[R91, A96] tended to aggravate CUS-induced anxiety-like behavior. Chronic stress increased the production of peripheral and central proinflammatory cytokines and induced the activation of microglia in the prelimbic cortex (PrL), which was blocked by MBP87-99[A91, A96]. Immunization with MBP-derived altered peptide ligands also rescued chronic stress-induced deficits in p11, phosphorylated cyclic adenosine monophosphate response element binding protein, and brain-derived neurotrophic factor expression. Moreover, microinjections of recombinant proinflammatory cytokines and the knockdown of p11 in the PrL blunted the antidepressant-like behavioral response to MBP87-99[A91, A96]. Altogether, these findings indicate that immunization with altered MBP peptide produces prolonged antidepressant-like effects in rats, and the behavioral response is mediated by inflammatory factors (particularly interleukin-6), and p11 signaling in the PrL. Immune-neural interactions may impact central nervous system function and alter an individual's response to stress.
Collapse
|
65
|
Vai B, Parenti L, Bollettini I, Cara C, Verga C, Melloni E, Mazza E, Poletti S, Colombo C, Benedetti F. Predicting differential diagnosis between bipolar and unipolar depression with multiple kernel learning on multimodal structural neuroimaging. Eur Neuropsychopharmacol 2020; 34:28-38. [PMID: 32238313 DOI: 10.1016/j.euroneuro.2020.03.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/24/2020] [Accepted: 03/06/2020] [Indexed: 01/10/2023]
Abstract
One of the greatest challenges in providing early effective treatment in mood disorders is the early differential diagnosis between major depression (MDD) and bipolar disorder (BD). A remarkable need exists to identify reliable biomarkers for these disorders. We integrate structural neuroimaging techniques (i.e. Tract-based Spatial Statistics, TBSS, and Voxel-based morphometry) in a multiple kernel learning procedure in order to define a predictive function of BD against MDD diagnosis in a sample of 148 patients. We achieved a balanced accuracy of 73.65% with a sensitivity for BD of 74.32% and specificity for MDD of 72.97%. Mass-univariates analyses showed reduced grey matter volume in right hippocampus, amygdala, parahippocampal, fusiform gyrus, insula, rolandic and frontal operculum and cerebellum, in BD compared to MDD. Volumes in these regions and in anterior cingulate cortex were also reduced in BD compared to healthy controls (n = 74). TBSS analyses revealed widespread significant effects of diagnosis on fractional anisotropy, axial, radial, and mean diffusivity in several white matter tracts, suggesting disruption of white matter microstructure in depressed patients compared to healthy controls, with worse pattern for MDD. To best of our knowledge, this is the first study combining grey matter and diffusion tensor imaging in predicting BD and MDD diagnosis. Our results prompt brain quantitative biomarkers and multiple kernel learning as promising tool for personalized treatment in mood disorders.
Collapse
Affiliation(s)
- Benedetta Vai
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy; Fondazione Centro San Raffaele, Milano, Italy.
| | - Lorenzo Parenti
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Irene Bollettini
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Cristina Cara
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Chiara Verga
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Elisa Melloni
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Elena Mazza
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Sara Poletti
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Cristina Colombo
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| | - Francesco Benedetti
- Division of Neuroscience, Psychiatry and Clinical Psychobiology Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy; University Vita-Salute San Raffaele, Milano, Italy
| |
Collapse
|
66
|
Single-nucleus transcriptomics of the prefrontal cortex in major depressive disorder implicates oligodendrocyte precursor cells and excitatory neurons. Nat Neurosci 2020; 23:771-781. [DOI: 10.1038/s41593-020-0621-y] [Citation(s) in RCA: 290] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 03/12/2020] [Indexed: 02/06/2023]
|
67
|
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
|
68
|
Reveals of New Candidate Active Components in Hemerocallis Radix and Its Anti-Depression Action of Mechanism Based on Network Pharmacology Approach. Int J Mol Sci 2020; 21:ijms21051868. [PMID: 32182911 PMCID: PMC7084327 DOI: 10.3390/ijms21051868] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/01/2020] [Accepted: 03/06/2020] [Indexed: 12/25/2022] Open
Abstract
The global depression population is showing a significant increase. Hemerocallis fulva L. is a common Traditional Chinese Medicine (TCM). Its flower buds are known to have ability to clear away heat and dampness, detoxify, and relieve depression. Ancient TCM literature shows that its roots have a beneficial effect in calming the spirit and even the temper in order to reduce the feeling of melancholy. Therefore, it is inferred that the root of Hemerocallis fulva L. can be used as a therapeutic medicine for depression. This study aims to uncover the pharmacological mechanism of the antidepressant effect of Hemerocallis Radix (HR) through network pharmacology method. During the analysis, 11 active components were obtained and screened using ADME—absorption, distribution, metabolism, and excretion— method. Furthermore, 267 HR targets and 740 depressive disorder (DD) targets were gathered from various databases. Then protein–protein interaction (PPI) network of HR and DD targets were constructed and cluster analysis was applied to further explore the connection between the targets. In addition, gene ontology (GO) enrichment and pathway analysis was applied to further verify that the biological process related to the target protein is associated with the occurrence of depression disorder. In conclusion, the most important bioactive components—anthraquinone, kaempferol, and vanillic acid—can alleviate depression symptoms by regulating MAOA, MAOB, and ESR1. The proposed network pharmacology strategy provides an integrating method to explore the therapeutic mechanism of multi-component drugs on a systematic level.
Collapse
|
69
|
Cuellar-Barboza AB, Sánchez-Ruiz JA, Rodriguez-Sanchez IP, González S, Calvo G, Lugo J, Costilla-Esquivel A, Martínez LE, Ibarra-Ramirez M. Gene expression in peripheral blood in treatment-free major depression. Acta Neuropsychiatr 2020; 32:1-10. [PMID: 32039744 DOI: 10.1017/neu.2020.5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Peripheral gene expression of several molecular pathways has been studied in major depressive disorder (MDD) with promising results. We sought to investigate some of these genes in a treatment-free Latino sample of Mexican descent. MATERIAL AND METHODS The sample consisted of 50 MDD treatment-free cases and 50 sex and age-matched controls. Gene expression of candidate genes of neuroplasticity (BDNF, p11, and VGF), inflammation (IL1A, IL1B, IL4, IL6, IL7, IL8, IL10, MIF, and TNFA), the canonical Wnt signaling pathway (TCF7L2, APC, and GSK3B), and mTOR, was compared in cases and controls. RNA was obtained from blood samples. We used bivariate analyses to compare subjects versus control mean mRNA quantification of target genes and lineal regression modelling to test for effects of age and body mass index on gene expression. RESULTS Most subjects were female (66%) with a mean age of 26.7 (SD 7.9) years. Only GSK3B was differentially expressed between cases and controls at a statistically significant level (p = 0.048). TCF7L-2 showed the highest number of correlations with MDD-related traits, yet these were modest in size. DISCUSSION GSK3B encodes a moderator of the canonical Wnt signaling pathway. It has a role in neuroplasticity, neuroprotection, depression, and other psychiatric phenotypes. We found that adding population diversity has the potential to elicit distinct peripheral gene expression markers in MDD and MDD-related traits. However, our results should only be considered as hypothesis-generating research that merits further replication in larger cohorts of similar ancestry.
Collapse
Affiliation(s)
- Alfredo B Cuellar-Barboza
- Department of Psychiatry, University Hospital, Universidad Autónoma de Nuevo León, Monterrey, México
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Jorge A Sánchez-Ruiz
- Department of Psychiatry, University Hospital, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Iram P Rodriguez-Sanchez
- Molecular and Structural Physiology Laboratory, School of Biological Sciences, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Sarai González
- Department of Psychiatry, University Hospital, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Geovana Calvo
- Department of Genetics, University Hospital, Universidad Autónoma de Nuevo León, Monterrey, México
| | - José Lugo
- Department of Genetics, University Hospital, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Antonio Costilla-Esquivel
- Department of Psychiatry, University Hospital, Universidad Autónoma de Nuevo León, Monterrey, México
- Centro de Investigación en Matemáticas A.C. (CIMAT), Monterrey, México
| | - Laura E Martínez
- Department of Genetics, University Hospital, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Marisol Ibarra-Ramirez
- Department of Genetics, University Hospital, Universidad Autónoma de Nuevo León, Monterrey, México
| |
Collapse
|
70
|
Fang X, Chen Y, Wang Y, Ren J, Zhang C. Depressive symptoms in schizophrenia patients: A possible relationship between SIRT1 and BDNF. Prog Neuropsychopharmacol Biol Psychiatry 2019; 95:109673. [PMID: 31247244 DOI: 10.1016/j.pnpbp.2019.109673] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 06/04/2019] [Accepted: 06/17/2019] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Depressive symptoms are commonly seen in schizophrenia. Increasing evidence implicates that both SIRT1 and BDNF closely related to the development of depression. So we here aimed to explore the effect of BDNF and SIRT1 on the depressive symptoms, and also explore the risk factors for the depressive symptoms in schizophrenia patients. METHODS A group of 203 participants (case/controls, 174/29) was recruited in the present work. Significant depression was classified by the CDSS score 7 or above. The psychotic symptoms and cognitive functions in schizophrenia patients were evaluated by PANSS and RBANS respectively. And the plasma concentration of SIRT1 and BDNF were measured in 167 participants (case/controls, 138/29). RESULTS Compared to healthy subjects, schizophrenia patients exhibited notably lower levels of BDNF (P < 0.05). And we observed that patients with depression displayed a worse psychotic symptom (P < 0.01), a poorer cognitive function(P < 0.05), as well as lower plasma BDNF and SIRT1 levels (Bonferroni corrected P < 0.05) compared to those no depressive patients. And the SIRT1 levels were positively correlated with BDNF levels in the case group (P < 0.001). And the regression analysis showed that negative symptoms and general psychopathology in PANSS, attention and delayed memory in RBANS, BDNF and SIRT1 were independent risk factors for depressive symptoms in schizophrenia. CONCLUSION Aside from the association with psychotic symptoms, we provided evidence suggesting that low BDNF and SIRT1 concentration value may be responsible for the occurrence of depression in schizophrenic patients.
Collapse
Affiliation(s)
- Xinyu Fang
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yan Chen
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yewei Wang
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Juanjuan Ren
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Chen Zhang
- Schizophrenia Program, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
71
|
Longitudinal transcriptome-wide gene expression analysis of sleep deprivation treatment shows involvement of circadian genes and immune pathways. Transl Psychiatry 2019; 9:343. [PMID: 31852885 PMCID: PMC6920477 DOI: 10.1038/s41398-019-0671-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 11/13/2019] [Accepted: 11/22/2019] [Indexed: 01/08/2023] Open
Abstract
Therapeutic sleep deprivation (SD) rapidly induces robust, transient antidepressant effects in a large proportion of major mood disorder patients suffering from a depressive episode, but underlying biological factors remain poorly understood. Research suggests that these patients may have altered circadian molecular genetic 'clocks' and that SD functions through 'resetting' dysregulated genes; additional factors may be involved, warranting further investigation. Leveraging advances in microarray technology enabling the transcriptome-wide assessment of gene expression, this study aimed to examine gene expression changes accompanying SD and recovery sleep in patients suffering from an episode of depression. Patients (N = 78) and controls (N = 15) underwent SD, with blood taken at the same time of day before SD, after one night of SD and after recovery sleep. A transcriptome-wide gene-by-gene approach was used, with a targeted look also taken at circadian genes. Furthermore, gene set enrichment, and longitudinal gene set analyses including the time point after recovery sleep, were conducted. Circadian genes were significantly affected by SD, with patterns suggesting that molecular clocks of responders and non-responders, as well as patients and controls respond differently to chronobiologic stimuli. Notably, gene set analyses revealed a strong widespread effect of SD on pathways involved in immune function and inflammatory response, such as those involved in cytokine and especially in interleukin signalling. Longitudinal gene set analyses showed that in responders these pathways were upregulated after SD; in non-responders, little response was observed. Our findings emphasize the close relationship between circadian, immune and sleep systems and their link to etiology of depression at the transcriptomic level.
Collapse
|
72
|
Liu W, Yan H, Zhou D, Cai X, Zhang Y, Li S, Li H, Li S, Zhou DS, Li X, Zhang C, Sun Y, Dai JP, Zhong J, Yao YG, Luo XJ, Fang Y, Zhang D, Ma Y, Yue W, Li M, Xiao X. The depression GWAS risk allele predicts smaller cerebellar gray matter volume and reduced SIRT1 mRNA expression in Chinese population. Transl Psychiatry 2019; 9:333. [PMID: 31819045 PMCID: PMC6901563 DOI: 10.1038/s41398-019-0675-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/19/2019] [Accepted: 11/27/2019] [Indexed: 12/28/2022] Open
Abstract
Major depressive disorder (MDD) is recognized as a primary cause of disability worldwide, and effective management of this illness has been a great challenge. While genetic component is supposed to play pivotal roles in MDD pathogenesis, the genetic and phenotypic heterogeneity of the illness has hampered the discovery of its genetic determinants. In this study, in an independent Han Chinese sample (1824 MDD cases and 3031 controls), we conducted replication analyses of two genetic loci highlighted in a previous Chinese MDD genome-wide association study (GWAS), and confirmed the significant association of a single nucleotide polymorphism (SNP) rs12415800 near SIRT1. Subsequently, using hypothesis-free whole-brain analysis in two independent Han Chinese imaging samples, we found that individuals carrying the MDD risk allele of rs12415800 exhibited aberrant gray matter volume in the left posterior cerebellar lobe compared with those carrying the non-risk allele. Besides, in independent Han Chinese postmortem brain and peripheral blood samples, the MDD risk allele of rs12415800 predicted lower SIRT1 mRNA levels, which was consistent with the reduced expression of this gene in MDD patients compared with healthy subjects. These results provide further evidence for the involvement of SIRT1 in MDD, and suggest that this gene might participate in the illness via affecting the development of cerebellum, a brain region that is potentially underestimated in previous MDD studies.
Collapse
Affiliation(s)
- Weipeng Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Hao Yan
- Peking University Sixth Hospital/Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health (Peking University) and National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Danyang Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xin Cai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yuyanan Zhang
- Peking University Sixth Hospital/Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health (Peking University) and National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
| | - Shiyi Li
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Huijuan Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shiwu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Dong-Sheng Zhou
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang, China
| | - Xingxing Li
- Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang, China
| | - Chen Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Sun
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei, China
- Chinese Brain Bank Center, Wuhan, Hubei, China
| | - Jia-Pei Dai
- Wuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei, China
- Chinese Brain Bank Center, Wuhan, Hubei, China
| | - Jingmei Zhong
- Psychiatry Department, The first people's hospital of Yunnan province, Kunming, Yunnan, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan, China
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, Yunnan, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yiru Fang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Dai Zhang
- Peking University Sixth Hospital/Institute of Mental Health, Beijing, China
- NHC Key Laboratory of Mental Health (Peking University) and National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China
- Peking-Tsinghua Joint Center for Life Sciences and PKU IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Yina Ma
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Weihua Yue
- Peking University Sixth Hospital/Institute of Mental Health, Beijing, China.
- NHC Key Laboratory of Mental Health (Peking University) and National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing, China.
- Peking-Tsinghua Joint Center for Life Sciences and PKU IDG/McGovern Institute for Brain Research, Peking University, Beijing, China.
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
73
|
Bhandage AK, Cunningham JL, Jin Z, Shen Q, Bongiovanni S, Korol SV, Syk M, Kamali-Moghaddam M, Ekselius L, Birnir B. Depression, GABA, and Age Correlate with Plasma Levels of Inflammatory Markers. Int J Mol Sci 2019; 20:ijms20246172. [PMID: 31817800 PMCID: PMC6941074 DOI: 10.3390/ijms20246172] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/17/2022] Open
Abstract
Immunomodulation is increasingly being recognised as a part of mental diseases. Here, we examined whether levels of immunological protein markers changed with depression, age, or the inhibitory neurotransmitter gamma-aminobutyric acid (GABA). An analysis of plasma samples from patients with a major depressive episode and control blood donors (CBD) revealed the expression of 67 inflammatory markers. Thirteen of these markers displayed augmented levels in patients compared to CBD. Twenty-one markers correlated with the age of the patients, whereas 10 markers correlated with the age of CBD. Interestingly, CST5 and CDCP1 showed the strongest correlation with age in the patients and CBD, respectively. IL-18 was the only marker that correlated with the MADRS-S scores of the patients. Neuronal growth factors (NGFs) were significantly enhanced in plasma from the patients, as was the average plasma GABA concentration. GABA modulated the release of seven cytokines in anti-CD3-stimulated peripheral blood mononuclear cells (PBMCs) from the patients. The study reveals significant changes in the plasma composition of small molecules during depression and identifies potential peripheral biomarkers of the disease.
Collapse
Affiliation(s)
- Amol K. Bhandage
- Department of Neuroscience, Physiology, Uppsala University, BMC, Box 593, 75124 Uppsala, Sweden; (A.K.B.); (Z.J.); (S.V.K.)
| | - Janet L. Cunningham
- Department of Neuroscience, Psychiatry, Uppsala University, 75185 Uppsala, Sweden; (J.L.C.); (S.B.); (M.S.); (L.E.)
| | - Zhe Jin
- Department of Neuroscience, Physiology, Uppsala University, BMC, Box 593, 75124 Uppsala, Sweden; (A.K.B.); (Z.J.); (S.V.K.)
| | - Qiujin Shen
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, 75108 Uppsala, Sweden; (Q.S.); (M.K.-M.)
| | - Santiago Bongiovanni
- Department of Neuroscience, Psychiatry, Uppsala University, 75185 Uppsala, Sweden; (J.L.C.); (S.B.); (M.S.); (L.E.)
| | - Sergiy V. Korol
- Department of Neuroscience, Physiology, Uppsala University, BMC, Box 593, 75124 Uppsala, Sweden; (A.K.B.); (Z.J.); (S.V.K.)
| | - Mikaela Syk
- Department of Neuroscience, Psychiatry, Uppsala University, 75185 Uppsala, Sweden; (J.L.C.); (S.B.); (M.S.); (L.E.)
| | - Masood Kamali-Moghaddam
- Department of Immunology, Genetics and Pathology, Science for Life laboratory, Uppsala University, 75108 Uppsala, Sweden; (Q.S.); (M.K.-M.)
| | - Lisa Ekselius
- Department of Neuroscience, Psychiatry, Uppsala University, 75185 Uppsala, Sweden; (J.L.C.); (S.B.); (M.S.); (L.E.)
| | - Bryndis Birnir
- Department of Neuroscience, Physiology, Uppsala University, BMC, Box 593, 75124 Uppsala, Sweden; (A.K.B.); (Z.J.); (S.V.K.)
- Correspondence: ; Tel.: +46-18-471-4622
| |
Collapse
|
74
|
Lapato DM, Roberson-Nay R, Kirkpatrick RM, Webb BT, York TP, Kinser PA. DNA methylation associated with postpartum depressive symptoms overlaps findings from a genome-wide association meta-analysis of depression. Clin Epigenetics 2019; 11:169. [PMID: 31779682 PMCID: PMC6883636 DOI: 10.1186/s13148-019-0769-z] [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: 04/08/2019] [Accepted: 10/22/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Perinatal depressive symptoms have been linked to adverse maternal and infant health outcomes. The etiology associated with perinatal depressive psychopathology is poorly understood, but accumulating evidence suggests that understanding inter-individual differences in DNA methylation (DNAm) patterning may provide insight regarding the genomic regions salient to the risk liability of perinatal depressive psychopathology. RESULTS Genome-wide DNAm was measured in maternal peripheral blood using the Infinium MethylationEPIC microarray. Ninety-two participants (46% African-American) had DNAm samples that passed all quality control metrics, and all participants were within 7 months of delivery. Linear models were constructed to identify differentially methylated sites and regions, and permutation testing was utilized to assess significance. Differentially methylated regions (DMRs) were defined as genomic regions of consistent DNAm change with at least two probes within 1 kb of each other. Maternal age, current smoking status, estimated cell-type proportions, ancestry-relevant principal components, days since delivery, and chip position served as covariates to adjust for technical and biological factors. Current postpartum depressive symptoms were measured using the Edinburgh Postnatal Depression Scale. Ninety-eight DMRs were significant (false discovery rate < 5%) and overlapped 92 genes. Three of the regions overlap loci from the latest Psychiatric Genomics Consortium meta-analysis of depression. CONCLUSIONS Many of the genes identified in this analysis corroborate previous allelic, transcriptomic, and DNAm association results related to depressive phenotypes. Future work should integrate data from multi-omic platforms to understand the functional relevance of these DMRs and refine DNAm association results by limiting phenotypic heterogeneity and clarifying if DNAm differences relate to the timing of onset, severity, duration of perinatal mental health outcomes of the current pregnancy or to previous history of depressive psychopathology.
Collapse
Affiliation(s)
- Dana M Lapato
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA. .,Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA.
| | - Roxann Roberson-Nay
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA.,Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Robert M Kirkpatrick
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA.,Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Bradley T Webb
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA.,Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Timothy P York
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA.,Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA.,Department of Obstetrics and Gynecology, Virginia Commonwealth University, Richmond, VA, USA
| | - Patricia A Kinser
- School of Nursing, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
75
|
Cook IA, Congdon E, Krantz DE, Hunter AM, Coppola G, Hamilton SP, Leuchter AF. Time Course of Changes in Peripheral Blood Gene Expression During Medication Treatment for Major Depressive Disorder. Front Genet 2019; 10:870. [PMID: 31620172 PMCID: PMC6760033 DOI: 10.3389/fgene.2019.00870] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 08/20/2019] [Indexed: 12/11/2022] Open
Abstract
Changes in gene expression (GE) during antidepressant treatment may increase understanding of the action of antidepressant medications and serve as biomarkers of efficacy. GE changes in peripheral blood are desirable because they can be assessed easily on multiple occasions during treatment. We report here on GE changes in 68 individuals who were treated for 8 weeks with either escitalopram alone, or escitalopram followed by bupropion. GE changes were assessed after 1, 2, and 8 weeks of treatment, with significant changes observed in 156, 121, and 585 peripheral blood gene transcripts, respectively. Thirty-one transcript changes were shared between the 1- and 8-week time points (seven upregulated, 24 downregulated). Differences were detected between the escitalopram- and bupropion-treated subjects, although there was no significant association between GE changes and clinical outcome. A subset of 18 genes overlapped with those previously identified as differentially expressed in subjects with MDD compared with healthy control subjects. There was statistically significant overlap between genes differentially expressed in the current and previous studies, with 10 genes overlapping in at least two previous studies. There was no enrichment for genes overexpressed in nervous system cell types, but there was a trend toward enrichment for genes in the WNT/β-catenin pathway in the anterior thalamus; three genes in this pathway showed differential expression in the present and in three previous studies. Our dataset and other similar studies will provide an important source of information about potential biomarkers of recovery and for potential dysregulation of GE in MDD.
Collapse
Affiliation(s)
- Ian A Cook
- Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Bioengineering, Henry Samueli School of Engineering at Applied Science, University of California, Los Angeles, Los Angeles, CA, United States
| | - Eliza Congdon
- Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - David E Krantz
- Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Aimee M Hunter
- Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Giovanni Coppola
- Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Steven P Hamilton
- Department of Psychiatry, Kaiser Permanente Northern California, San Francisco, CA, United States.,Department of Psychiatry, University of California, San Francisco, San Francisco, CA, United States
| | - Andrew F Leuchter
- Neuromodulation Division, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
76
|
Vennu V, Misra H, Misra A. Depressive symptoms and the risk of arthritis: A survival analysis using data from the osteoarthritis initiative. Indian J Psychiatry 2019; 61:444-450. [PMID: 31579152 PMCID: PMC6767814 DOI: 10.4103/psychiatry.indianjpsychiatry_241_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Some studies investigated the association between depressive symptoms and arthritis; however, no longitudinal studies have documented the relationship between developing depressive symptoms and the risk of arthritis. Therefore, this study evaluated whether the development of depressive symptoms was associated with an elevated risk of arthritis. MATERIALS AND METHODS A survival analysis using Cox regression models was applied to osteoarthritis initiative data obtained over 6 years from adults (n = 3,662) aged ≥45 years at baseline. Developing depressive symptoms was defined using the 20-item Center for Epidemiologic Studies Depression scale (cutoff 16 points) between baseline and 1 year. Arthritis was defined answering "yes" to the following self-reported question: "Did the doctor say you developed arthritis since the last clinic visit about 1 year ago?" over the 6-year follow-up period. RESULTS The hazard ratios for developing arthritis were 3.51 (95% confidence interval [CI] = 2.32-5.29) and 2.03 (95% CI = 1.45-2.85) for men and women, respectively, as compared to those who did not develop depressive symptoms. There was a significantly (χ 2 = 73.672, P < 0.0001) lower survival probability at each time point throughout the study among men and women who developed depressive symptoms. CONCLUSION In both men and women, developing depressive symptoms increased the risk of arthritis, and the survival probability decreased at each time point.
Collapse
Affiliation(s)
- Vishal Vennu
- Department of Pharmacy, School of Pharmacy, Lingaya's University, Faridabad, Haryana, India.,Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Harsh Misra
- Department of Pharmacology, Mulayam Singh Yadav Medical College and Hospital, Meerut, Uttar Pradesh, India
| | - Asha Misra
- Department of OBS and Gynaecology, Mulayam Singh Yadav Medical College and Hospital, Meerut, Uttar Pradesh, India
| |
Collapse
|
77
|
de Kluiver H, Jansen R, Milaneschi Y, Penninx BWJH. Involvement of inflammatory gene expression pathways in depressed patients with hyperphagia. Transl Psychiatry 2019; 9:193. [PMID: 31431611 PMCID: PMC6702221 DOI: 10.1038/s41398-019-0528-0] [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: 10/24/2018] [Revised: 04/25/2019] [Accepted: 06/20/2019] [Indexed: 12/21/2022] Open
Abstract
The pathophysiology of major depressive disorder (MDD) is highly heterogeneous. Previous evidence at the DNA level as well as on the serum protein level suggests that the role of inflammation in MDD pathology is stronger in patients with hyperphagia during an active episode. Which inflammatory pathways differ in MDD patients with hyperphagia inflammatory pathways in terms of gene expression is unknown. We analyzed whole-blood gene expression profiles of 881 current MDD cases and 331 controls from the Netherlands Study of Depression and Anxiety (NESDA). The MDD patients were stratified according to patients with hyperphagia (characterized by increased appetite and/or weight, N = 246) or hypophagia (characterized by decreased appetite and/or weight, N = 342). Using results of differential gene expression analysis between controls and the MDD subgroups, enrichment of curated inflammatory pathways was estimated. The majority of the pathways were significantly (FDR < 0.1) enriched in the expression profiles of MDD cases with hyperphagia, including top pathways related to factors responsible for the onset of inflammatory response ('caspase', 'GATA3', 'NFAT', and 'inflammasomes' pathways). Only two pathways ('adaptive immune system' and 'IL-8- and CXCR2-mediated signaling') were enriched in the MDD with hypophagia subgroup, these were also enriched in the total current MDD group and the group with hyperphagia. This confirms the importance of inflammation in MDD pathology of patients with hyperphagia, and suggests that distinguishing more uniform MDD phenotypes can help in finding their pathophysiological basis.
Collapse
Affiliation(s)
- Hilde de Kluiver
- Amsterdam UMC, Vrije Universiteit, Department of Psychiatry, Amsterdam Public Health research institute and Amsterdam Neuroscience, Oldenaller 1, 1081 HJ, Amsterdam, the Netherlands.
| | - Rick Jansen
- grid.484519.5Amsterdam UMC, Vrije Universiteit, Department of Psychiatry, Amsterdam Neuroscience, Oldenaller 1, 1081 HJ Amsterdam, the Netherlands
| | - Yuri Milaneschi
- 0000 0004 0435 165Xgrid.16872.3aAmsterdam UMC, Vrije Universiteit, Department of Psychiatry, Amsterdam Public Health research institute and Amsterdam Neuroscience, Oldenaller 1, 1081 HJ Amsterdam, the Netherlands
| | - Brenda W. J. H. Penninx
- 0000 0004 0435 165Xgrid.16872.3aAmsterdam UMC, Vrije Universiteit, Department of Psychiatry, Amsterdam Public Health research institute and Amsterdam Neuroscience, Oldenaller 1, 1081 HJ Amsterdam, the Netherlands
| |
Collapse
|
78
|
Esquivel-Rendón E, Vargas-Mireles J, Cuevas-Olguín R, Miranda-Morales M, Acosta-Mares P, García-Oscos F, Pineda JC, Salgado H, Rose-John S, Atzori M. Interleukin 6 Dependent Synaptic Plasticity in a Social Defeat-Susceptible Prefrontal Cortex Circuit. Neuroscience 2019; 414:280-296. [DOI: 10.1016/j.neuroscience.2019.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/08/2019] [Accepted: 07/01/2019] [Indexed: 12/29/2022]
|
79
|
Li H, Chang H, Song X, Liu W, Li L, Wang L, Yang Y, Zhang L, Li W, Zhang Y, Zhou DS, Li X, Zhang C, Fang Y, Sun Y, Dai JP, Luo XJ, Yao YG, Xiao X, Lv L, Li M. Integrative analyses of major histocompatibility complex loci in the genome-wide association studies of major depressive disorder. Neuropsychopharmacology 2019; 44:1552-1561. [PMID: 30771788 PMCID: PMC6785001 DOI: 10.1038/s41386-019-0346-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/01/2019] [Accepted: 02/12/2019] [Indexed: 11/09/2022]
Abstract
Recent European genome-wide association studies (GWAS) have revealed strong statistical correlations between MDD and numerous zero-to-high linked variants in the genomic region containing major histocompatibility complex (MHC) genes (MHC region), but the underlying biological mechanisms are still unclear. To better understand the roles of this genomic region in the neurobiology of MDD, we applied a convergent functional genomics approach to integrate GWAS data of MDD relevant biological phenotypes, gene-expression analyses results obtained from brain samples, and genetic analyses of independent Chinese MDD samples. We observed that independent MDD risk variants in the MHC region were also significantly associated with the relevant biological phenotypes in the predicted directions, including the emotional and cognitive-related phenotypes. Gene-expression analyses further revealed that mRNA expression levels of several MHC region genes in the human brain were associated with MDD risk SNPs and diagnostic status. For instance, a brain-enriched gene ZNF603P consistently showed lower mRNA levels in the individuals carrying MDD risk alleles and in MDD patients. Remarkably, we further found that independent MDD risk SNPs in the MHC region likely converged to affect the mRNA level(s) of the same gene(s), and Europeans and Han Chinese populations have a substantial shared genetic and molecular basis underlying MDD risk associations in the MHC region. These results highlighted several potential pivotal genes at the MHC region in the pathogenesis of MDD. Their common impacts on multiple psychiatric relevant phenotypes also implicated the neurological processes shared by different psychological processes, such as mood and/or cognition, shedding lights on their potential biological mechanisms.
Collapse
Affiliation(s)
- Huijuan Li
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China ,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan China
| | - Hong Chang
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China
| | - Xueqin Song
- grid.412633.1The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Weipeng Liu
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China ,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan China
| | - Lingyi Li
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China
| | - Lu Wang
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China
| | - Yongfeng Yang
- 0000 0004 1808 322Xgrid.412990.7Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China ,0000 0004 1808 322Xgrid.412990.7Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China
| | - Luwen Zhang
- 0000 0004 1808 322Xgrid.412990.7Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China ,0000 0004 1808 322Xgrid.412990.7Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China
| | - Wenqiang Li
- 0000 0004 1808 322Xgrid.412990.7Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China ,0000 0004 1808 322Xgrid.412990.7Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China
| | - Yan Zhang
- 0000 0004 1808 322Xgrid.412990.7Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan China ,0000 0004 1808 322Xgrid.412990.7Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan China
| | - Dong-Sheng Zhou
- 0000 0004 1782 599Xgrid.452715.0Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang China
| | - Xingxing Li
- 0000 0004 1782 599Xgrid.452715.0Department of Psychiatry, Ningbo Kangning Hospital, Ningbo, Zhejiang China
| | - Chen Zhang
- 0000 0004 0368 8293grid.16821.3cShanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiru Fang
- 0000 0004 0368 8293grid.16821.3cShanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Sun
- 0000 0000 9147 9053grid.412692.aWuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei China ,Chinese Brain Bank Center, Wuhan, Hubei China
| | - Jia-Pei Dai
- 0000 0000 9147 9053grid.412692.aWuhan Institute for Neuroscience and Neuroengineering, South-Central University for Nationalities, Wuhan, Hubei China ,Chinese Brain Bank Center, Wuhan, Hubei China
| | - Xiong-Jian Luo
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China ,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan China ,0000000119573309grid.9227.eCenter for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan China
| | - Yong-Gang Yao
- 0000000119573309grid.9227.eKey Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan China ,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan China ,0000000119573309grid.9227.eCAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Luxian Lv
- Department of Psychiatry, Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China. .,Henan Key Lab of Biological Psychiatry, Xinxiang Medical University, Xinxiang, Henan, China. .,Henan Province People's Hospital, Zhengzhou, Henan, China.
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China. .,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
80
|
Kato TA, Katsuki R, Kubo H, Shimokawa N, Sato-Kasai M, Hayakawa K, Kuwano N, Umene-Nakano W, Tateno M, Setoyama D, Kang D, Watabe M, Sakamoto S, Teo AR, Kanba S. Development and validation of the 22-item Tarumi's Modern-Type Depression Trait Scale: Avoidance of Social Roles, Complaint, and Low Self-Esteem (TACS-22). Psychiatry Clin Neurosci 2019; 73:448-457. [PMID: 30900331 PMCID: PMC6850625 DOI: 10.1111/pcn.12842] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/13/2019] [Accepted: 03/19/2019] [Indexed: 12/12/2022]
Abstract
AIM Understanding premorbid personality is important, especially when considering treatment selection. Historically, the premorbid personality of patients with major depression in Japan was described as Shuchaku-kishitsu [similar to Typus melancholicus], as proposed by Shimoda in the 1930s. Since around 2000, there have been increased reports in Japan of young adults with depression who have had premorbid personality differing from the traditional type. In 2005, Tarumi termed this novel condition 'dysthymic-type depression,' and more recently the condition has been called Shin-gata/Gendai-gata Utsu-byo [modern-type depression (MTD)]. We recently developed a semi-structured diagnostic interview to evaluate MTD. Development of a tool that enables understanding of premorbid personality in a short time, especially at the early stage of treatment, is desirable. The object of this study was to develop a self-report scale to evaluate the traits of MTD, and to assess the scale's psychometric properties, diagnostic accuracy, and biological validity. METHODS A sample of 340 participants from clinical and community settings completed measures. Psychometric properties were assessed with factor analysis. Diagnostic accuracy of the MTD traits was compared against a semi-structured interview. RESULTS The questionnaire contained 22 items across three subscales, thus we termed it the 22-item Tarumi's Modern-Type Depression Trait Scale: Avoidance of Social Roles, Complaint, and Low Self-Esteem (TACS-22). Internal consistency, test-retest reliability, and convergent validity were all satisfactory. Among patients with major depression, the area under the curve was 0.757 (sensitivity of 63.1% and specificity of 82.9%) and the score was positively correlated with plasma tryptophan. CONCLUSION The TACS-22 possessed adequate psychometric properties and diagnostic accuracy in an initial sample of Japanese adults. Additional research on its ability to support clinical assessment of MTD is warranted.
Collapse
Affiliation(s)
- Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryoko Katsuki
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroaki Kubo
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Norihiro Shimokawa
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mina Sato-Kasai
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Hayakawa
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobuki Kuwano
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Wakako Umene-Nakano
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaru Tateno
- Tokiwa Child Development Center, Tokiwa Hospital, Sapporo, Japan.,Department of Neuropsychiatry, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Daiki Setoyama
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Motoki Watabe
- School of Business, Monash University, Bandar Sunway, Malaysia
| | - Shinji Sakamoto
- Department of Psychology, College of Humanities and Sciences, Nihon University, Tokyo, Japan
| | - Alan R Teo
- VA Portland Health Care System, HSR&D Center to Improve Veteran Involvement in Care, Portland, USA.,Department of Psychiatry, Oregon Health & Science University, Portland, USA.,School of Public Health, Oregon Health & Science University, Portland, USA.,School of Public Health, Portland State University, Portland, USA
| | - Shigenobu Kanba
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
81
|
Lee CH, Giuliani F. The Role of Inflammation in Depression and Fatigue. Front Immunol 2019; 10:1696. [PMID: 31379879 PMCID: PMC6658985 DOI: 10.3389/fimmu.2019.01696] [Citation(s) in RCA: 367] [Impact Index Per Article: 61.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022] Open
Abstract
Depression and fatigue are conditions responsible for heavy global societal burden, especially in patients already suffering from chronic diseases. These symptoms have been identified by those affected as some of the most disabling symptoms which affect the quality of life and productivity of the individual. While many factors play a role in the development of depression and fatigue, both have been associated with increased inflammatory activation of the immune system affecting both the periphery and the central nervous system (CNS). This is further supported by the well-described association between diseases that involve immune activation and these symptoms in autoimmune disorders, such as multiple sclerosis and immune system activation in response to infections, like sepsis. Treatments for depression also support this immunopsychiatric link. Antidepressants have been shown to decrease inflammation, while higher levels of baseline inflammation predict lower treatment efficacy for most treatments. Those patients with higher initial immune activation may on the other hand be more responsive to treatments targeting immune pathways, which have been found to be effective in treating depression and fatigue in some cases. These results show strong support for the hypothesis that depression and fatigue are associated with an increased activation of the immune system which may serve as a valid target for treatment. Further studies should focus on the pathways involved in these symptoms and the development of treatments that target those pathways will help us to better understand these conditions and devise more targeted treatments.
Collapse
Affiliation(s)
- Chieh-Hsin Lee
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Fabrizio Giuliani
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.,Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
82
|
Gerring ZF, Gamazon ER, Derks EM, for the Major Depressive Disorder Working Group of the Psychiatric Genomics Consortium. A gene co-expression network-based analysis of multiple brain tissues reveals novel genes and molecular pathways underlying major depression. PLoS Genet 2019; 15:e1008245. [PMID: 31306407 PMCID: PMC6658115 DOI: 10.1371/journal.pgen.1008245] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 07/25/2019] [Accepted: 06/12/2019] [Indexed: 11/30/2022] Open
Abstract
Major depression is a common and severe psychiatric disorder with a highly polygenic genetic architecture. Genome-wide association studies have successfully identified multiple independent genetic loci that harbour variants associated with major depression, but the exact causal genes and biological mechanisms are largely unknown. Tissue-specific network approaches may identify molecular mechanisms underlying major depression and provide a biological substrate for integrative analyses. We provide a framework for the identification of individual risk genes and gene co-expression networks using genome-wide association summary statistics and gene expression information across multiple human brain tissues and whole blood. We developed a novel gene-based method called eMAGMA that leverages tissue-specific eQTL information to identify 99 biologically plausible risk genes associated with major depression, of which 58 are novel. Among these novel associations is Complement Factor 4A (C4A), recently implicated in schizophrenia through its role in synaptic pruning during postnatal development. Major depression risk genes were enriched in gene co-expression modules in multiple brain tissues and the implicated gene modules contained genes involved in synaptic signalling, neuronal development, and cell transport pathways. Modules enriched with major depression signals were strongly preserved across brain tissues, but were weakly preserved in whole blood, highlighting the importance of using disease-relevant tissues in genetic studies of psychiatric traits. We identified tissue-specific genes and gene co-expression networks associated with major depression. Our novel analytical framework can be used to gain fundamental insights into the functioning of the nervous system in major depression and other brain-related traits. Although genome-wide association studies have identified genetic risk variants associated with major depression, our understanding of the mechanisms through which they influence disease susceptibility remains largely unknown. Genetic risk variants are highly enriched in non-coding regions of the genome and affect gene expression. Genes are co-expressed and regulate the activity of one another to form highly organized networks. In this study, we generate tissue-specific gene co-expression networks, each containing groups of functionally related genes or “modules”, to delineate gene co-expression and thereby facilitate the identification of gene processes in major depression. We developed and applied a novel research methodology (called “eMAGMA”) which integrates genetic and transcriptomic information in a tissue-specific analysis to identify risk genes and test for their enrichment in gene co-expression modules. Using this novel approach, we identified gene modules in multiple tissues that are both enriched with major depression genetic association signals and biologically meaningful pathways. We also show the disease implicated gene modules are strongly preserved across brain regions, but not in whole blood, suggesting unique patterns of gene co-expression within the two tissue types. Our novel analytical framework provides important insights into the functional genetics major depression and can be applied to other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Zachary F. Gerring
- Translational Neurogenomics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- * E-mail:
| | - Eric R. Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Clare Hall, University of Cambridge, Cambridge, United Kingdom
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Eske M. Derks
- Translational Neurogenomics Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | |
Collapse
|
83
|
Liu W, Zhang L, Zheng D, Zhang Y. Umbilical cord blood-based gene signatures related to prenatal major depressive disorder. Medicine (Baltimore) 2019; 98:e16373. [PMID: 31305436 PMCID: PMC6641773 DOI: 10.1097/md.0000000000016373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Prenatal exposure to depression has been considered as a risk factor for adverse childhood, while it is accompanied by unknown molecular mechanisms. The aim of this study was to identify differentially expressed genes (DEGs) and associated biological processes between cord blood samples from neonates born to mothers who exposed to major depressive disorder (MDD) and healthy mothers. METHODS The microarray data GSE114852 were downloaded to analyze the mRNA expression profiles of umbilical cord blood with 31 samples exposed to prenatal MDD and 62 samples with healthy mothers. Kyoto Encyclopedia of Genes and Genomes pathway and Gene ontology enrichment analyses were conducted to identify associated biochemical pathways and functional categories of the DEGs. The protein-protein interaction network was constructed and the top 10 hub genes in the network were predicted. RESULTS The results showed several immunity related processes, such as "phagosome", "Epstein-Barr virus infection", "proteasome", "positive regulation of I-kappaB kinase/NF-kappaB signaling", "interferon-gamma-mediated signaling pathway", and "tumor necrosis factor" presented significant differences between two groups. Most of the hub genes (for example PSMD2, PSMD6, PSMB8, PSMB9) were also associated with immune pathways. CONCLUSION This bioinformatic analysis demonstrated immune-mediated mechanisms might play a fatal role in abnormalities in fetal gene expression profiles caused by prenatal MDD.
Collapse
Affiliation(s)
- Wenhua Liu
- Department of Psychology and Mental Health, Huaihe Hospital of Henan University, Kaifeng City, Henan Province
| | - Lan Zhang
- Department of Psychology and Mental Health, Second Affiliated Hospital of Lanzhou University, Lanzhou City, Gansu Province
| | | | - Yijie Zhang
- Department of Respiratory and Critical Care Medicine, Huaihe Hospital of Henan University, Kaifeng City, Henan Province, China
| |
Collapse
|
84
|
Leirer DJ, Iyegbe CO, Di Forti M, Patel H, Carra E, Fraietta S, Colizzi M, Mondelli V, Quattrone D, Lally J, Ajnakina O, Lee SH, Curtis CJ, Breen G, Pariante C, Aitchison K, Dazzan P, Murray RM, Dobson RJB, Newhouse SJ. Differential gene expression analysis in blood of first episode psychosis patients. Schizophr Res 2019; 209:88-97. [PMID: 31113746 PMCID: PMC6677921 DOI: 10.1016/j.schres.2019.05.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 02/22/2019] [Accepted: 05/05/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Psychosis is a condition influenced by an interaction of environmental and genetic factors. Gene expression studies can capture these interactions; however, studies are usually performed in patients who are in remission. This study uses blood of first episode psychosis patients, in order to characterise deregulated pathways associated with psychosis symptom dimensions. METHODS Peripheral blood from 149 healthy controls and 131 first episode psychosis patients was profiled using Illumina HT-12 microarrays. A case/control differential expression analysis was performed, followed by correlation of gene expression with positive and negative syndrome scale (PANSS) scores. Enrichment analyses were performed on the associated gene lists. We test for pathway differences between first episode psychosis patients who qualify for a Schizophrenia diagnosis against those who do not. RESULTS A total of 978 genes were differentially expressed and enriched for pathways associated to immune function and the mitochondria. Using PANSS scores we found that positive symptom severity was correlated with immune function, while negative symptoms correlated with mitochondrial pathways. CONCLUSIONS Our results identified gene expression changes correlated with symptom severity and showed that key pathways are modulated by positive and negative symptom dimensions.
Collapse
Affiliation(s)
- Daniel J Leirer
- MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, Box P080, De Crespigny Park, SE5 8AF London, UK.
| | - Conrad O Iyegbe
- Department of Psychosis Studies, Institute of Psychiatry, Kings College London, Box P092, De Crespigny Park, SE5 8AF London, UK
| | - Marta Di Forti
- MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, Box P080, De Crespigny Park, SE5 8AF London, UK
| | - Hamel Patel
- MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, Box P080, De Crespigny Park, SE5 8AF London, UK; Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, Box P080, De Crespigny Park, SE5 8AF London, UK
| | - Elena Carra
- Department of Psychosis Studies, Institute of Psychiatry, Kings College London, Box P092, De Crespigny Park, SE5 8AF London, UK
| | - Sara Fraietta
- Department of Psychosis Studies, Institute of Psychiatry, Kings College London, Box P092, De Crespigny Park, SE5 8AF London, UK
| | - Marco Colizzi
- Department of Psychosis Studies, Institute of Psychiatry, Kings College London, Box P092, De Crespigny Park, SE5 8AF London, UK
| | - Valeria Mondelli
- Department of Psychological Medicine, Institute of Psychiatry, Kings College London, De Crespigny Park, SE5 8AF London, UK
| | - Diego Quattrone
- MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, Box P080, De Crespigny Park, SE5 8AF London, UK
| | - John Lally
- Department of Psychosis Studies, Institute of Psychiatry, Kings College London, Box P092, De Crespigny Park, SE5 8AF London, UK
| | - Olesya Ajnakina
- Department of Psychosis Studies, Institute of Psychiatry, Kings College London, Box P092, De Crespigny Park, SE5 8AF London, UK
| | - Sang Hyuck Lee
- MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, Box P080, De Crespigny Park, SE5 8AF London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London
| | - Charles J Curtis
- MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, Box P080, De Crespigny Park, SE5 8AF London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London
| | - Gerome Breen
- MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, Box P080, De Crespigny Park, SE5 8AF London, UK; NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London
| | - Carmine Pariante
- Department of Psychological Medicine, Institute of Psychiatry, Kings College London, De Crespigny Park, SE5 8AF London, UK
| | - Katherine Aitchison
- Departments of Psychiatry and Medical Genetics, University of Alberta, 11361-87 Avenue, AB T6G 2E1, Edmonton, Canada
| | - Paola Dazzan
- MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, Box P080, De Crespigny Park, SE5 8AF London, UK; Department of Psychological Medicine, Institute of Psychiatry, Kings College London, De Crespigny Park, SE5 8AF London, UK
| | - Robin M Murray
- Department of Psychosis Studies, Institute of Psychiatry, Kings College London, Box P092, De Crespigny Park, SE5 8AF London, UK
| | - Richard J B Dobson
- NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM) & Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London; Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, Box P080, De Crespigny Park, SE5 8AF London, UK
| | - Stephen J Newhouse
- MRC Social, Genetic and Developmental Psychiatry (SGDP) Centre, Institute of Psychiatry, Psychology and Neuroscience, Box P080, De Crespigny Park, SE5 8AF London, UK; Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, Box P080, De Crespigny Park, SE5 8AF London, UK
| |
Collapse
|
85
|
Zhang X, Abdellaoui A, Rucker J, de Jong S, Potash JB, Weissman MM, Shi J, Knowles JA, Pato C, Pato M, Sobell J, Smit JH, Hottenga JJ, de Geus EJ, Lewis CM, Buttenschøn HN, Craddock N, Jones I, Jones L, McGuffin P, Mors O, Owen MJ, Preisig M, Rietschel M, Rice JP, Rivera M, Uher R, Gejman PV, Sanders AR, Boomsma D, Penninx BWJH, Breen G, Levinson DF. Genome-wide Burden of Rare Short Deletions Is Enriched in Major Depressive Disorder in Four Cohorts. Biol Psychiatry 2019; 85:1065-1073. [PMID: 31003785 PMCID: PMC6750266 DOI: 10.1016/j.biopsych.2019.02.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/29/2019] [Accepted: 02/19/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is moderately heritable, with a high prevalence and a presumed high heterogeneity. Copy number variants (CNVs) could contribute to the heritable component of risk, but the two previous genome-wide association studies of rare CNVs did not report significant findings. METHODS In this meta-analysis of four cohorts (5780 patients and 6626 control subjects), we analyzed the association of MDD to 1) genome-wide burden of rare deletions and duplications, partitioned by length (<100 kb or >100 kb) and other characteristics, and 2) individual rare exonic CNVs and CNV regions. RESULTS Patients with MDD carried significantly more short deletions than control subjects (p = .0059) but not long deletions or short or long duplications. The confidence interval for long deletions overlapped with that for short deletions, but long deletions were 70% less frequent genome-wide, reducing the power to detect increased burden. The increased burden of short deletions was primarily in intergenic regions. Short deletions in cases were also modestly enriched for high-confidence enhancer regions. No individual CNV achieved thresholds for suggestive or significant association after genome-wide correction. p values < .01 were observed for 15q11.2 duplications (TUBGCP5, CYFIP1, NIPA1, and NIPA2), deletions in or near PRKN or MSR1, and exonic duplications of ATG5. CONCLUSIONS The increased burden of short deletions in patients with MDD suggests that rare CNVs increase the risk of MDD by disrupting regulatory regions. Results for longer deletions were less clear, but no large effects were observed for long multigenic CNVs (as seen in schizophrenia and autism). Further studies with larger sample sizes are warranted.
Collapse
Affiliation(s)
- Xianglong Zhang
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, USA.,Department of Genetics, Stanford University School of Medicine, USA
| | - Abdel Abdellaoui
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, the Netherlands.,Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - James Rucker
- The Institute of Psychiatry, King’s College London, UK
| | | | - James B. Potash
- Department of Psychiatry, Johns Hopkins University School of Medicine, USA
| | - Myrna M. Weissman
- Department of Psychiatry, Columbia University, and New York State Psychiatric Institute, USA
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, Biostatistics Branch, National Cancer Institute, USA
| | - James A. Knowles
- Department of Cell Biology, Downstate Medical Center College of Medicine, USA
| | - Carlos Pato
- Downstate Medical Center College of Medicine, USA
| | - Michele Pato
- Department of Psychiatry, Downstate Medical Center College of Medicine, USA
| | - Janet Sobell
- Department of Psychiatry and Behavioral Sciences, University of Southern California, USA
| | - Johannes H. Smit
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Psychiatry, Amsterdam Public Health and GGz inGeest, Amsterdam, the Netherlands
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, the Netherlands
| | - Eco J.C. de Geus
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, the Netherlands
| | - Cathryn M Lewis
- MRC Social Genetic and Developmental Psychiatry Centre, King’s College London, London, UK.,Department of Medical & Molecular Genetics, King’s College London, London, UK
| | - Henriette N Buttenschøn
- Department of Clinical Medicine, Translational Neuropsychiatry Unit, Aarhus University, Aarhus, DK.,iSEQ, Centre for Integrative Sequencing, Aarhus University, Aarhus, DK
| | - Nick Craddock
- Department of Psychological Medicine, Cardiff University, Cardiff, UK
| | - Ian Jones
- Department of Psychological Medicine, Cardiff University, Cardiff, UK
| | - Lisa Jones
- Institute of Health and Society, University of Worcester, Worcester, UK
| | - Peter McGuffin
- MRC Social Genetic and Developmental Psychiatry Centre, King’s College London, London, UK
| | - Ole Mors
- Psychosis Research Unit, Aarhus University Hospital, Risskov, Aarhus, DK
| | - Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
| | - Martin Preisig
- Department of Psychiatry, University Hospital of Lausanne, Prilly, Switzerland
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - John P Rice
- Department of Psychiatry, Washington University in Saint Louis School of Medicine, Saint Louis, USA
| | - Margarita Rivera
- MRC Social Genetic and Developmental Psychiatry Centre, King’s College London, London, UK.,Department of Biochemistry and Molecular Biology II, Institute of Neurosciences, Center for Biomedical Research, University of Granada, Granada
| | - Rudolf Uher
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Pablo V. Gejman
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Chicago, USA.,Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, USA
| | - Alan R. Sanders
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Chicago, USA.,Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, USA
| | - Dorret Boomsma
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, the Netherlands
| | - Brenda W. J. H. Penninx
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Psychiatry, Amsterdam Public Health and GGz inGeest, Amsterdam, the Netherlands
| | - Gerome Breen
- MRC Social Genetic and Developmental Psychiatry Centre, King’s College London, London, UK.,NIHR BRC for Mental Health, King’s College London, London, UK
| | - Douglas F. Levinson
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, USA
| |
Collapse
|
86
|
Gamazon ER, Zwinderman AH, Cox NJ, Denys D, Derks EM. Multi-tissue transcriptome analyses identify genetic mechanisms underlying neuropsychiatric traits. Nat Genet 2019; 51:933-940. [PMID: 31086352 PMCID: PMC6590703 DOI: 10.1038/s41588-019-0409-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 04/03/2019] [Indexed: 01/02/2023]
Abstract
The genetic architecture of psychiatric disorders is characterized by a large number of small-effect variants1 located primarily in non-coding regions, suggesting that the underlying causal effects may influence disease risk by modulating gene expression2-4. We provide comprehensive analyses using transcriptome data from an unprecedented collection of tissues to gain pathophysiological insights into the role of the brain, neuroendocrine factors (adrenal gland) and gastrointestinal systems (colon) in psychiatric disorders. In each tissue, we perform PrediXcan analysis and identify trait-associated genes for schizophrenia (n associations = 499; n unique genes = 275), bipolar disorder (n associations = 17; n unique genes = 13), attention deficit hyperactivity disorder (n associations = 19; n unique genes = 12) and broad depression (n associations = 41; n unique genes = 31). Importantly, both PrediXcan and summary-data-based Mendelian randomization/heterogeneity in dependent instruments analyses suggest potentially causal genes in non-brain tissues, showing the utility of these tissues for mapping psychiatric disease genetic predisposition. Our analyses further highlight the importance of joint tissue approaches as 76% of the genes were detected only in difficult-to-acquire tissues.
Collapse
Affiliation(s)
- Eric R Gamazon
- Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
- Data Science Institute, Vanderbilt University, Nashville, TN, USA.
- Clare Hall, University of Cambridge, Cambridge, UK.
| | - Aeilko H Zwinderman
- Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Nancy J Cox
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Damiaan Denys
- Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Eske M Derks
- Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
- QIMR Berghofer, Translational Neurogenomics Group, Brisbane, Queensland, Australia.
| |
Collapse
|
87
|
Pariante CM. Did Spider-Man Work in the NESDA Cohort? In Immunopsychiatry, With Great Power Comes Great Responsibility. Biol Psychiatry 2019; 85:787-788. [PMID: 31046936 DOI: 10.1016/j.biopsych.2019.03.980] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/25/2019] [Accepted: 03/25/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Carmine M Pariante
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.
| |
Collapse
|
88
|
Lamers F, Milaneschi Y, Smit JH, Schoevers RA, Wittenberg G, Penninx BWJH. Longitudinal Association Between Depression and Inflammatory Markers: Results From the Netherlands Study of Depression and Anxiety. Biol Psychiatry 2019; 85:829-837. [PMID: 30819515 DOI: 10.1016/j.biopsych.2018.12.020] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/20/2018] [Accepted: 12/31/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND While cross-sectional associations of inflammatory markers interleukin-6 (IL-6) and C-reactive protein with major depressive disorder are well established, evidence for longitudinal associations mostly comes from studies on depression symptoms, not diagnoses. This study examined cross-sectional and bidirectional longitudinal associations between depression diagnosis and symptoms in an adult sample over a 6-year period. METHODS Data were obtained from the baseline (n = 2416) and 2- and 6-year follow-up assessments (n = 1925 and n = 1924, respectively) of the Netherlands Study of Depression and Anxiety. C-reactive protein and IL-6 were assessed at each wave, as were the Composite International Diagnostic Interview and Inventory of Depressive Symptomatology. Linear mixed models and generalized estimating equation models with a binomial distribution were used to study longitudinal associations between depression and inflammation and vice versa. RESULTS There was a consistent cross-sectional association between current depressive disorder (vs. no current disorder) and symptoms with IL-6 across all follow-up measurements (Cohen's ddepression diagnosis = 0.06, p = .017; Bstandardized Inventory of Depressive Symptomatology = 0.029, SE = 0.011, p = .008). In longitudinal analyses, higher IL-6 levels predicted subsequent chronic course in those with a diagnosis at baseline in women but not in men (odds ratiowomen = 1.13, 95% confidence interval = 1.04-1.23), and both depressive disorder and high severity predicted higher IL-6 levels at the subsequent follow-up (p values < .01). In contrast, C-reactive protein was not associated with current depression in cross-sectional and longitudinal analyses. CONCLUSIONS In this longitudinal study, cross-sectional and bidirectional longitudinal associations were found between depression and IL-6 levels. This underlines the importance of targeting inflammation pathways in the treatment of major depressive disorder. IL-6 could be a potential marker for patient profiling in personalized medicine approaches.
Collapse
Affiliation(s)
- Femke Lamers
- Department of Psychiatry, Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Johannes H Smit
- Department of Psychiatry, Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Robert A Schoevers
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
89
|
Zhang L, Zhang X, Zheng J, Liu Y, Wang J, Wang G, Zhang HP, Kang DY, Peng ZG, Ji YL, Wang L, Gibson PG, Wang G. Depressive symptom-associated IL-1β and TNF-α release correlates with impaired bronchodilator response and neutrophilic airway inflammation in asthma. Clin Exp Allergy 2019; 49:770-780. [PMID: 30675924 DOI: 10.1111/cea.13346] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 10/16/2018] [Accepted: 12/20/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Depressive symptoms worsen asthma outcomes; however, the mechanism remains largely unexplored. OBJECTIVE This study aimed to determine whether depressive symptom-associated immune inflammation correlates with impaired bronchodilator response (BDR) and airway inflammatory phenotypes. METHODS Eligible adults with asthma (n = 198) underwent clinical assessment, sputum induction and blood sampling. Depressive symptoms were defined by scores on the depression subscale of the Hospital Anxiety and Depression Scale (HADS-D). Pre- and post-bronchodilator spirometry was performed for BDR. Airway inflammatory phenotypes were defined by sputum cell counts. CRP, IL-1β, IL-5, IL-6, IL-8, TNF-α, IFN-γ, CCL17 and CCL22 in serum and sputum were detected. RESULTS Compared with the non-depressive group (n = 174), the depressive group (n = 24) exhibited impaired BDR (P = 0.032) and increased sputum neutrophils (P = 0.023), which correlated with the HADS-D scores (P = 0.027 and P = 0.029). Levels of IL-1β, TNF-α and IFN-γ in the serum and those of IL-1β and IFN-γ in the sputum were elevated in the depressive group compared to those in the non-depressive group (all P < 0.05). Multiple regression models indicated that TNF-α in the sputum and IL-1β, IL-6 and IFN-γ in both the serum and sputum were inversely associated with BDR; TNF-α in the sputum and IL-1β in both the serum and sputum were positively correlated with sputum neutrophils. Mediation analyses revealed that IL-1β and TNF-α in the sputum and IL-1β in both the serum and sputum mediate the correlations of the HADS-D scores with BDR and sputum neutrophils, respectively. CONCLUSIONS AND CLINICAL RELEVANCE Asthma patients with depressive symptoms present worse asthma control, which is most likely explained by impaired BDR and neutrophilic airway inflammation. IL-1β and TNF-α, which are two key pro-inflammatory cytokines that mediate the correlation of depressive symptoms with impaired BDR and neutrophilic airway inflammation, may serve as targeted biomarkers in the neuropsychological phenotype of asthma; however, this result needs to be further validated.
Collapse
Affiliation(s)
- Li Zhang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Zhang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China.,Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Zheng
- Department of Integrated Traditional Chinese and Western Medicine, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Ying Liu
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ji Wang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Wang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Ping Zhang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - De Ying Kang
- Department of Evidence-based Medicine and Clinical Epidemiology, West China Hospital, Sichuan University, Chengdu, China
| | - Zu Gui Peng
- Department of Psychiatry, The Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Lin Ji
- Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Wang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Peter Gerard Gibson
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Hunter Medical Research Institute, Priority Research Centre for Healthy Lungs, University of Newcastle, Newcastle, New South Wales, Australia
| | - Gang Wang
- Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Pneumology Group, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China.,Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
90
|
Chimienti F, Cavarec L, Vincent L, Salvetat N, Arango V, Underwood MD, Mann JJ, Pujol JF, Weissmann D. Brain region-specific alterations of RNA editing in PDE8A mRNA in suicide decedents. Transl Psychiatry 2019; 9:91. [PMID: 30770787 PMCID: PMC6377659 DOI: 10.1038/s41398-018-0331-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 10/26/2018] [Accepted: 11/13/2018] [Indexed: 12/20/2022] Open
Abstract
Phosphodiesterases (PDE) are key modulators of signal transduction and are involved in inflammatory cell activation, memory and cognition. There is a two-fold decrease in the expression of phosphodiesterase 8A (PDE8A) in the temporal cortex of major depressive disorder (MDD) patients. Here, we studied PDE8A mRNA-editing profile in two architectonically distinct neocortical regions in a clinically well-characterized cohort of age- and sex-matched non-psychiatric drug-free controls and depressed suicide decedents. By using capillary electrophoresis single-stranded conformational polymorphism (CE-SSCP), a previously validated technique to identify A-to-I RNA modifications, we report the full editing profile of PDE8A in the brain, including identification of two novel editing sites. Editing of PDE8A mRNA displayed clear regional difference when comparing dorsolateral prefrontal cortex (BA9) and anterior cingulate cortex (BA24). Furthermore, we report significant intra-regional differences between non-psychiatric control individuals and depressed suicide decedents, which could discriminate the two populations. Taken together, our results (i) highlight the importance of immune/inflammatory markers in major depressive disorder and suicide and (ii) establish a direct relationship between A-to-I RNA modifications of peripheral markers and A-to-I RNA editing-related modifications in brain. This work provides the first immune response-related brain marker for suicide and could pave the way for the identification of a blood-based biomarker that predicts suicidal behavior.
Collapse
Affiliation(s)
- Fabrice Chimienti
- ALCEDIAG/ Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Montpellier, France.
| | - Laurent Cavarec
- grid.465535.4Genomic Vision, Green Square, 80-84 rue des Meuniers, 92220 Bagneux, France
| | - Laurent Vincent
- grid.457349.8Commissariat à l’Energie Atomique, Fontenay aux Roses, France
| | - Nicolas Salvetat
- ALCEDIAG/ Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Montpellier, France
| | - Victoria Arango
- 0000 0000 8499 1112grid.413734.6Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY USA ,0000000419368729grid.21729.3fDepartment of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY USA
| | - Mark D. Underwood
- 0000 0000 8499 1112grid.413734.6Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY USA ,0000000419368729grid.21729.3fDepartment of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY USA
| | - J. John Mann
- 0000 0000 8499 1112grid.413734.6Division of Molecular Imaging and Neuropathology, New York State Psychiatric Institute, New York, NY USA ,0000000419368729grid.21729.3fDepartment of Psychiatry, Columbia University College of Physicians and Surgeons, New York, NY USA
| | | | - Dinah Weissmann
- ALCEDIAG/ Sys2Diag, CNRS UMR 9005, Parc Euromédecine, Montpellier, France
| |
Collapse
|
91
|
Altered mRNA expressions for N-methyl-D-aspartate receptor-related genes in WBC of patients with major depressive disorder. J Affect Disord 2019; 245:1119-1125. [PMID: 30699855 DOI: 10.1016/j.jad.2018.12.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/27/2018] [Accepted: 12/08/2018] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Major depressive disorder (MDD) is a complex mental disorder. The lack of well-established biomarkers hinders its diagnosis, treatment, and new-drug development. N-methyl-D-aspartate receptor (NMDAR) dysfunction has been implicated in the pathogenesis of MDD. This study examined whether expressions of the NMDAR-related genes are characteristic of MDD. METHODS Expressions of NMDAR-related genes including SRR, SHMT2, PSAT1, GCAT, GAD1, SLC1A4, NRG1 and COMT in peripheral WBCs of 110 patients with MDD (25 drug-naïve, 21 drug-free, and 64 medicated patients) and 125 healthy individuals were measured using quantitative PCR. RESULTS The mRNA expression levels of SRR, PSAT1, GCAT, GAD1, NRG1 and COMT were significantly different among the four groups (all p < 0.05). For drug-naïve patients, the ΔΔCT values of SRR, PSAT1, GCAT, GAD1, and NRG1 mRNA expressions were significantly different from those in healthy individuals (all p < 0.05). The ROC analysis of the ΔΔCT values of the target genes for differentiating drug-naïve patients from healthy controls showed an excellent sensitivity (0.960) and modest specificity (0.640) (AUC = 0.889). Drug-free and medicated patients obtained less favorable AUC values while compared to healthy controls. The results for the age- and sex-matched cohort were similar to those of the unmatched cohort. CONCLUSIONS This is the first study demonstrating that the peripheral mRNA expression levels of NMDAR-related genes may be altered in patients with MDD, especially drug-naïve individuals. The finding supports the NMDAR hypothesis of depression. Whether mRNA expresssion of NMDAR-related genes could serve as a potential biomarker of MDD deserves further investigations.
Collapse
|
92
|
Savell KE, Bach SV, Zipperly ME, Revanna JS, Goska NA, Tuscher JJ, Duke CG, Sultan FA, Burke JN, Williams D, Ianov L, Day JJ. A Neuron-Optimized CRISPR/dCas9 Activation System for Robust and Specific Gene Regulation. eNeuro 2019; 6:ENEURO.0495-18.2019. [PMID: 30863790 PMCID: PMC6412672 DOI: 10.1523/eneuro.0495-18.2019] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/21/2019] [Accepted: 01/27/2019] [Indexed: 12/19/2022] Open
Abstract
CRISPR-based technology has provided new avenues to interrogate gene function, but difficulties in transgene expression in post-mitotic neurons has delayed incorporation of these tools in the central nervous system (CNS). Here, we demonstrate a highly efficient, neuron-optimized dual lentiviral CRISPR-based transcriptional activation (CRISPRa) system capable of robust, modular, and tunable gene induction and multiplexed gene regulation across several primary rodent neuron culture systems. CRISPRa targeting unique promoters in the complex multi-transcript gene brain-derived neurotrophic factor (Bdnf) revealed both transcript- and genome-level selectivity of this approach, in addition to highlighting downstream transcriptional and physiological consequences of Bdnf regulation. Finally, we illustrate that CRISPRa is highly efficient in vivo, resulting in increased protein levels of a target gene in diverse brain structures. Taken together, these results demonstrate that CRISPRa is an efficient and selective method to study gene expression programs in brain health and disease.
Collapse
Affiliation(s)
- Katherine E. Savell
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Svitlana V. Bach
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Morgan E. Zipperly
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Jasmin S. Revanna
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Nicholas A. Goska
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Jennifer J. Tuscher
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Corey G. Duke
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Faraz A. Sultan
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Julia N. Burke
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Derek Williams
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Lara Ianov
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, 35294
| | - Jeremy J. Day
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, 35294
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, 35294
| |
Collapse
|
93
|
Mustafin RN, Enikeeva RF, Davydova YD, Khusnutdinova EK. The Role of Epigenetic Factors in the Development of Depressive Disorders. RUSS J GENET+ 2018. [DOI: 10.1134/s1022795418120104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
94
|
Ciobanu LG, Sachdev PS, Trollor JN, Reppermund S, Thalamuthu A, Mather KA, Cohen-Woods S, Stacey D, Toben C, Schubert KO, Baune BT. Co-expression network analysis of peripheral blood transcriptome identifies dysregulated protein processing in endoplasmic reticulum and immune response in recurrent MDD in older adults. J Psychiatr Res 2018; 107:19-27. [PMID: 30312913 DOI: 10.1016/j.jpsychires.2018.09.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/26/2018] [Accepted: 09/28/2018] [Indexed: 02/09/2023]
Abstract
The molecular factors involved in the pathophysiology of major depressive disorder (MDD) remain poorly understood. One approach to examine the molecular basis of MDD is co-expression network analysis, which facilitates the examination of complex interactions between expression levels of individual genes and how they influence biological pathways affected in MDD. Here, we applied an unsupervised gene-network based approach to a prospective experimental design using microarray genome-wide gene expression from the peripheral whole blood of older adults. We utilised the Sydney Memory and Ageing Study (sMAS, N = 521) and the Older Australian Twins Study (OATS, N = 186) as discovery and replication cohorts, respectively. We constructed networks using Weighted Gene Co-expression Network Analysis (WGCNA), and correlated identified modules with four subtypes of depression: single episode, current, recurrent, and lifetime MDD. Four modules of highly co-expressed genes were associated with recurrent MDD (N = 27) in our discovery cohort (FDR<0.2), with no significant findings for a single episode, current or lifetime MDD. Functional characterisation of these modules revealed a complex interplay between dysregulated protein processing in the endoplasmic reticulum (ER), and innate and adaptive immune response signalling, with possible involvement of pathogen-related pathways. We were underpowered to replicate findings at the network level in an independent cohort (OATS), however; we found a significant overlap for 9 individual genes with similar co-expression and dysregulation patterns associated with recurrent MDD in both cohorts. Overall, our findings support other reports on dysregulated immune response and protein processing in the ER in MDD and provide novel insights into the pathophysiology of depression.
Collapse
Affiliation(s)
- Liliana G Ciobanu
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, 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, University of Adelaide, South Australia, Australia
| | - K Oliver Schubert
- Discipline of Psychiatry, Adelaide Medical School, 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
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, South Australia, Australia; Department of Psychiatry, Melbourne Medical School, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville VIC, 3010, Australia.
| |
Collapse
|
95
|
Hasselmann H, Gamradt S, Taenzer A, Nowacki J, Zain R, Patas K, Ramien C, Paul F, Wingenfeld K, Piber D, Gold SM, Otte C. Pro-inflammatory Monocyte Phenotype and Cell-Specific Steroid Signaling Alterations in Unmedicated Patients With Major Depressive Disorder. Front Immunol 2018; 9:2693. [PMID: 30532752 PMCID: PMC6265986 DOI: 10.3389/fimmu.2018.02693] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/31/2018] [Indexed: 12/21/2022] Open
Abstract
Several lines of evidence have strongly implicated inflammatory processes in the pathobiology of major depressive disorder (MDD). However, the cellular origin of inflammatory signals and their specificity remain unclear. We examined the phenotype and glucocorticoid signaling in key cell populations of the innate immune system (monocytes) vs. adaptive immunity (T cells) in a sample of 35 well-characterized, antidepressant-free patients with MDD and 35 healthy controls individually matched for age, sex, smoking status and body mass index. Monocyte and T cell phenotype was assessed by flow cytometry. Cell-specific steroid signaling was determined by mRNA expression of pre-receptor regulation (11β-hydroxysteroid dehydrogenase type 1; 11β -HSD1), steroid receptor expression [glucocorticoid receptor (GR) and mineralocorticoid receptor (MR)], and the downstream target glucocorticoid-induced leucine-zipper (GILZ). We also collected salivary cortisol samples (8:00 a.m. and 10:00 p.m.) on two consecutive days. Patients showed a shift toward a pro-inflammatory phenotype characterized by higher frequency and higher absolute numbers of non-classical monocytes. No group differences were observed in major T cell subset frequencies and phenotype. Correspondingly, gene expression indicative of steroid resistance (i.e., lower expression of GR and GILZ) in patients with MDD was specific to monocytes and not observed in T cells. Monocyte phenotype and steroid receptor expression was not related to cortisol levels or serum levels of IL-6, IL-1β, or TNF-α. Our results thus suggest that in MDD, cells of the innate and adaptive immune system are differentially affected with shifts in monocyte subsets and lower expression of steroid signaling related genes.
Collapse
Affiliation(s)
- Helge Hasselmann
- Klinik für Psychiatrie und Psychotherapie, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefanie Gamradt
- Klinik für Psychiatrie und Psychotherapie, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Aline Taenzer
- Klinik für Psychiatrie und Psychotherapie, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jan Nowacki
- Klinik für Psychiatrie und Psychotherapie, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Rami Zain
- Klinik für Psychiatrie und Psychotherapie, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kostas Patas
- Institut für Neuroimmunologie und Multiple Sklerose, (INIMS), Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg Eppendorf, Hamburg, Germany
| | - Caren Ramien
- Institut für Neuroimmunologie und Multiple Sklerose, (INIMS), Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg Eppendorf, Hamburg, Germany
| | - Friedemann Paul
- NeuroCure Clinical Research Center, (NCRC), Charité - Universitätsmedizin Berlin, Berlin, Germany.,Experimental and Clinical Research Center, Max Delbrueck Center for Molecular Medicine and Charité Universitätsmedizin Berlin, Berlin, Germany.,Klinik für Neurologie, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Katja Wingenfeld
- Klinik für Psychiatrie und Psychotherapie, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dominique Piber
- Klinik für Psychiatrie und Psychotherapie, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan M Gold
- Klinik für Psychiatrie und Psychotherapie, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Institut für Neuroimmunologie und Multiple Sklerose, (INIMS), Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg Eppendorf, Hamburg, Germany.,Medizinische Klinik m.S. Psychosomatik, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Otte
- Klinik für Psychiatrie und Psychotherapie, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
96
|
Rey R, Chauvet-Gelinier JC, Suaud-Chagny MF, Ragot S, Bonin B, d'Amato T, Teyssier JR. Distinct Expression Pattern of Epigenetic Machinery Genes in Blood Leucocytes and Brain Cortex of Depressive Patients. Mol Neurobiol 2018; 56:4697-4707. [PMID: 30377985 PMCID: PMC6647377 DOI: 10.1007/s12035-018-1406-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/24/2018] [Indexed: 12/25/2022]
Abstract
In major depressive disorder (MDD), altered gene expression in brain cortex and blood leucocytes may be due to aberrant expression of epigenetic machinery coding genes. Here, we explore the expression of these genes both at the central and peripheral levels. Using real-time quantitative PCR technique, we first measured expression levels of genes encoding DNA and histone modifying enzymes in the dorsolateral prefrontal cortex (DLPFC) and cingulate cortex (CC) of MDD patients (n = 24) and healthy controls (n = 12). For each brain structure, transcripts levels were compared between subject groups. In an exploratory analysis, we then compared the candidate gene expressions between a subgroup of MDD patients with psychotic characteristics (n = 13) and the group of healthy subjects (n = 12). Finally, we compared transcript levels of the candidate genes in blood leucocytes between separate samples of MDD patients (n = 17) and healthy controls (n = 16). In brain and blood leucocytes of MDD patients, we identified an overexpression of genes encoding enzymes which transfer repressive transcriptional marks: HDAC4-5-6-8 and DNMT3B in the DLPFC, HDAC2 in the CC and blood leucocytes. In the DLPFC of patients with psychotic characteristics, two genes (KAT2A and UBE2A) were additionally overexpressed suggesting a shift to a more transcriptionally permissive conformation of chromatin. Aberrant activation of epigenetic repressive systems may be involved in MDD pathogenesis both in brain tissue and blood leucocytes.
Collapse
Affiliation(s)
- Romain Rey
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Psychiatric Disorders: from Resistance to Response Team, F-69000, Lyon, France. .,University Lyon 1, F-69000, Villeurbanne, France. .,Schizophrenia Expert Centre, Le Vinatier Hospital, Bron, France. .,INSERM U1028; CNRS UMR5292; Université Claude Bernard Lyon 1; Centre de Recherche en Neurosciences de Lyon, Equipe PSYR2; Centre Hospitalier Le Vinatier, Pole Est, Centre Expert Schizophrénie, 95 boulevard Pinel BP 30039, 69678, Bron Cedex, France.
| | - Jean-Christophe Chauvet-Gelinier
- Psychiatry Unit, Neurosciences Department, Le Bocage University Hospital, Marion Building, Dijon, France.,Laboratory of Psychopathology and Medical Psychology (IFR 100), Bourgogne University, Dijon, France
| | - Marie-Françoise Suaud-Chagny
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Psychiatric Disorders: from Resistance to Response Team, F-69000, Lyon, France.,University Lyon 1, F-69000, Villeurbanne, France.,Schizophrenia Expert Centre, Le Vinatier Hospital, Bron, France
| | - Sylviane Ragot
- Department of Genetics and Laboratory of Molecular Genetics, University Hospital, Dijon, France
| | - Bernard Bonin
- Psychiatry Unit, Neurosciences Department, Le Bocage University Hospital, Marion Building, Dijon, France.,Laboratory of Psychopathology and Medical Psychology (IFR 100), Bourgogne University, Dijon, France
| | - Thierry d'Amato
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Psychiatric Disorders: from Resistance to Response Team, F-69000, Lyon, France.,University Lyon 1, F-69000, Villeurbanne, France.,Schizophrenia Expert Centre, Le Vinatier Hospital, Bron, France
| | - Jean-Raymond Teyssier
- Department of Genetics and Laboratory of Molecular Genetics, University Hospital, Dijon, France
| |
Collapse
|
97
|
Fujita Y, Yamashita T. Sirtuins in Neuroendocrine Regulation and Neurological Diseases. Front Neurosci 2018; 12:778. [PMID: 30416425 PMCID: PMC6213750 DOI: 10.3389/fnins.2018.00778] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Silent information regulator 1 (SIRT1) is a mammalian homolog of the nicotinamide adenine dinucleotide (NAD)-dependent deacetylase sirtuin family. Sirtuin was originally studied as the lifespan-extending gene, silent information regulator 2 (SIRT2) in budding yeast. There are seven mammalian homologs of sirtuin (SIRT1–7), and SIRT1 is the closest homolog to SIRT2. SIRT1 modulates various key targets via deacetylation. In addition to histones, these targets include transcription factors, such as forkhead box O (FOXO), Ku70, p53, NF-κB, PPAR-gamma co-activator 1-alpha (PGC-1α), and peroxisome proliferator-activated receptor γ (PPARγ). SIRT1 has many biological functions, including aging, cell survival, differentiation, and metabolism. Genetic and physiological analyses in animal models have shown beneficial roles for SIRT1 in the brain during both development and adulthood. Evidence from in vivo and in vitro studies have revealed that SIRT1 regulates the cellular fate of neural progenitors, axon elongation, dendritic branching, synaptic plasticity, and endocrine function. In addition to its importance in physiological processes, SIRT1 has also been implicated in protection of neurons from degeneration in models of neurological diseases, such as traumatic brain injury and Alzheimer’s disease. In this review, we focus on the role of SIRT1 in the neuroendocrine system and neurodegenerative diseases. We also discuss the potential therapeutic implications of targeting the sirtuin pathway.
Collapse
Affiliation(s)
- Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan.,WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan.,WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.,Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
98
|
Zhang J, Yang J, Han D, Zhao X, Ma J, Ban B, Zhu X, Yang Y, Cao D, Qiu X. Dvl3 polymorphism interacts with life events and pro-inflammatory cytokines to influence major depressive disorder susceptibility. Sci Rep 2018; 8:14181. [PMID: 30242173 PMCID: PMC6155061 DOI: 10.1038/s41598-018-31530-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 07/18/2018] [Indexed: 12/16/2022] Open
Abstract
The purpose of this study is to explore Dvl3 variants and their interaction with negative life events on MDD susceptibility in a Chinese Han population. Additionally, we also attempted to identify whether there is an association between Dvl3 variants and pro-inflammatory cytokines. A total of 1102 participants, consisting of 550 patients with MDD and 552 healthy subjects, were recruited for genotyping by TaqMan allelic discrimination assay. Pro-inflammatory cytokine mRNA levels in peripheral blood were measured by QPCR. After the assessment of negative life events by the Life Events Scale, the Dvl3 gene–environment interaction (G × E) and risk factors were evaluated using generalized multifactor dimensionality reduction method (GMDR) and logistic regression analysis, respectively. This study is the first to reveal the interaction between Dvl3 allelic variations and negative life events as well as pro-inflammatory cytokines on MDD susceptibility in a Chinese Han population.
Collapse
Affiliation(s)
- Jian Zhang
- Psychology Department of the Public Health Institute of Harbin Medical University, Heilongjiang Province, Harbin, China
| | - Jiarun Yang
- Psychology Department of the Public Health Institute of Harbin Medical University, Heilongjiang Province, Harbin, China
| | - Dong Han
- Psychology Department of the Public Health Institute of Harbin Medical University, Heilongjiang Province, Harbin, China
| | - Xueyan Zhao
- Psychology Department of the Public Health Institute of Harbin Medical University, Heilongjiang Province, Harbin, China
| | - Jingsong Ma
- Psychology Department of the Public Health Institute of Harbin Medical University, Heilongjiang Province, Harbin, China
| | - Bo Ban
- Affiliated Hosptial of Jining Medical University, Shandong Province, Jining, China
| | - Xiongzhao Zhu
- Medical Psychological Institute of the Second Xiangya Hospital of Central South University, Hunan Province, Changsha, China
| | - Yanjie Yang
- Psychology Department of the Public Health Institute of Harbin Medical University, Heilongjiang Province, Harbin, China.
| | - Depin Cao
- Harbin Medical University, Heilongjiang Province, Harbin, China.
| | - Xiaohui Qiu
- Psychology Department of the Public Health Institute of Harbin Medical University, Heilongjiang Province, Harbin, China.
| |
Collapse
|
99
|
Hori H, Nakamura S, Yoshida F, Teraishi T, Sasayama D, Ota M, Hattori K, Kim Y, Higuchi T, Kunugi H. Integrated profiling of phenotype and blood transcriptome for stress vulnerability and depression. J Psychiatr Res 2018; 104:202-210. [PMID: 30103068 DOI: 10.1016/j.jpsychires.2018.08.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/03/2018] [Accepted: 08/02/2018] [Indexed: 12/17/2022]
Abstract
Etiology of depression and its vulnerability remains elusive. Using a latent profile analysis on dimensional personality traits, we previously identified 3 different phenotypes in the general population, namely stress-resilient, -vulnerable, and -resistant groups. Here we performed microarray-based blood gene expression profiling of these 3 groups (n = 20 for each group) in order to identify genes involved in stress vulnerability as it relates to the risk of depression. Identified differentially expressed genes among the groups were most markedly enriched in ribosome-related pathways. These ribosomal genes, which included ribosomal protein L17 (RPL17) and ribosomal protein L34 (RPL34), were upregulated in relation to the stress vulnerability. Protein-protein interaction and correlational co-expression analyses of the differentially expressed genes/non-coding RNAs consistently showed that functional networks involving ribosomes were affected. The significant upregulation of RPL17 and RPL34 was also observed in depressed patients compared to healthy controls, as confirmed in 2 independent case-control datasets by using pooled microarray data and qPCR experiments (total number of subjects was 122 and 166, respectively). Moreover, the upregulation of RPL17 and RPL34 was most marked in DSM-IV major depressive disorder, followed by in bipolar disorder, and then in schizophrenia, suggesting some diagnostic specificity of these markers as well as their general roles in stress vulnerability. These results suggest that ribosomal genes, particularly RPL17 and RPL34, can play integral roles in stress vulnerability and depression across nonclinical and clinical conditions. This study presents an opportunity to understand how multiple psychological traits and underlying molecular mechanisms interact to render individuals vulnerable to depression.
Collapse
Affiliation(s)
- Hiroaki Hori
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan; Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, 187-8553, Japan.
| | | | - Fuyuko Yoshida
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan
| | - Toshiya Teraishi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan
| | - Daimei Sasayama
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan
| | - Miho Ota
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan
| | - Kotaro Hattori
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan
| | - Yoshiharu Kim
- Department of Behavioral Medicine, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, 187-8553, Japan
| | - Teruhiko Higuchi
- National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, 187-8502, Japan.
| |
Collapse
|
100
|
Woo HI, Lim SW, Myung W, Kim DK, Lee SY. Differentially expressed genes related to major depressive disorder and antidepressant response: genome-wide gene expression analysis. Exp Mol Med 2018; 50:1-11. [PMID: 30076325 PMCID: PMC6076250 DOI: 10.1038/s12276-018-0123-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 03/25/2018] [Accepted: 04/17/2018] [Indexed: 12/15/2022] Open
Abstract
Treatment response to antidepressants is limited and varies among patients with major depressive disorder (MDD). To discover genes and mechanisms related to the pathophysiology of MDD and antidepressant treatment response, we performed gene expression analyses using peripheral blood specimens from 38 MDD patients and 14 healthy individuals at baseline and at 6 weeks after the initiation of either selective serotonin reuptake inhibitor (SSRI) or mirtazapine treatment. The results were compared with results from public microarray data. Seven differentially expressed genes (DEGs) between MDD patients and controls were identified in our study and in the public microarray data: CD58, CXCL8, EGF, TARP, TNFSF4, ZNF583, and ZNF587. CXCL8 was among the top 10 downregulated genes in both studies. Eight genes related to SSRI responsiveness, including BTNL8, showed alterations in gene expression in MDD. The expression of the FCRL6 gene differed between SSRI responders and nonresponders and changed after SSRI treatment compared to baseline. In evaluating the response to mirtazapine, 21 DEGs were identified when comparing MDD patients and controls and responders and nonresponders. These findings suggest that the pathophysiology of MDD and treatment response to antidepressants are associated with a number of processes, including DNA damage and apoptosis, that can be induced by immune activation and inflammation.
Collapse
Affiliation(s)
- Hye In Woo
- Department of Laboratory Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Shinn-Won Lim
- SAIHST, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea
| | - Woojae Myung
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Doh Kwan Kim
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Soo-Youn Lee
- Department of Clinical Pharmacology & Therapeutics, Samsung Medical Center, Seoul, Korea.
- Department of Laboratory Medicine & Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|