1
|
Giatti S, Cioffi L, Diviccaro S, Chrostek G, Piazza R, Melcangi RC. Transcriptomic Profile of the Male Rat Hypothalamus and Nucleus Accumbens After Paroxetine Treatment and Withdrawal: Possible Causes of Sexual Dysfunction. Mol Neurobiol 2025; 62:4935-4951. [PMID: 39495228 DOI: 10.1007/s12035-024-04592-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
Paroxetine, a selective serotonin reuptake inhibitor (SSRI), may induce sexual dysfunction during treatment and upon discontinuation. The mechanisms involved have been poorly explored so far. We have analyzed, by RNA sequencing, the whole transcriptomic profile in the hypothalamus and nucleus accumbens (NAc) (two brain regions involved in sexual behavior) of male rats daily treated for 2 weeks with paroxetine (T0) and at 1 month of withdrawal (T1). Data here reported show seven differentially expressed genes (DEGs) at T0 and 1 at T1 in the hypothalamus and 245 at T0 and 6 at T1 in the NAc. In addition, Gene-Set Enrichment, Gene Ontology, and Reactome analyses confirm that inflammatory signature and immune system activation were present at T0 in both brain areas. Considering that inflammation is generally associated with depression and that no paradigms inducing the pathology were here applied, these SSRI pro-depressive effects should be considered in patients without a clear indication of depression. Moreover, DEGs related to neurotransmitters with a role in sexual behavior and the reward system, such as dopamine (e.g., sialyltransferase 8B-ST8SIA3), glutamate (e.g., glutamate receptor ionotropic delta-2-GRID2) and GABA (e.g., glutamate decarboxylase type 2-GAD2) or associated with neurexin and neuroligin pathways and brain-derived neurotrophic factor (BDNF) signaling, were reported to be dysregulated in the NAc, further confirming dysfunction in this brain area. Interestingly, the analysis of DEGs altered at T1 in the NAc confirms the persistence of some of these side effects providing further information for post-SSRI sexual dysfunction (PSSD) etiopathogenesis.
Collapse
Affiliation(s)
- Silvia Giatti
- Dipartimento Di Scienze Farmacologiche E Biomolecolari, "Rodolfo Paoletti", Università Degli Studi Di Milano, Via Balzaretti 9, 20133, Milan, Italy.
| | - Lucia Cioffi
- Dipartimento Di Scienze Farmacologiche E Biomolecolari, "Rodolfo Paoletti", Università Degli Studi Di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Silvia Diviccaro
- Dipartimento Di Scienze Farmacologiche E Biomolecolari, "Rodolfo Paoletti", Università Degli Studi Di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Gabriela Chrostek
- Dipartimento Di Scienze Farmacologiche E Biomolecolari, "Rodolfo Paoletti", Università Degli Studi Di Milano, Via Balzaretti 9, 20133, Milan, Italy
| | - Rocco Piazza
- Dipartimento Di Medicina E Chirurgia, Università Di Milano-Bicocca, Milan, Italy
| | - Roberto Cosimo Melcangi
- Dipartimento Di Scienze Farmacologiche E Biomolecolari, "Rodolfo Paoletti", Università Degli Studi Di Milano, Via Balzaretti 9, 20133, Milan, Italy.
| |
Collapse
|
2
|
Savitz J, McKinney BA, Meier TB, Zheng H, Ford BN, Yolken RH, Teague TK, Cole SW. Nuclear factor kappa-B cell (NF-κB), interferon regulatory Factor, and glucocorticoid receptor pathway activation in major depressive Disorder: The role of cytomegalovirus infection. Brain Behav Immun 2025; 123:1052-1060. [PMID: 39532200 PMCID: PMC11624063 DOI: 10.1016/j.bbi.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/18/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
Altered activity of major immunoregulatory pathways has been reported in major depressive disorder (MDD) and is thought to underlie the elevations in circulating inflammatory mediators present in a subgroup of patients. However, the drivers of these changes in gene expression remain unclear. One potential modulator of immune function is viral infection. Here we examined the relationship between cytomegalovirus (CMV), a common herpesvirus, that has been shown to be a pathological cofactor in inflammatory disorders, and activity of key coordinators of the innate inflammatory response in MDD. We used RNAseq to characterize gene expression differences in in 79 unmedicated individuals with MDD and 80 healthy controls (HCs). A well-established bioinformatic strategy was used to quantify transcription control pathway activity based on the relative prevalence of pre-specified transcription factor-binding motifs in the promoters of differentially expressed genes. The main aim was to characterize diagnostic differences in immunoregulatory pathway activity and determine if these were related to CMV serostatus or antibody titer (viral reactivation). Significantly increased activity of interferon regulatory factor 1 (IRF1) and nuclear factor kappa-B cell (NF-κB) pathways was observed in the MDD group compared with HCs. Transcript Origin Analyses using cell-specific reference transcriptomes indicated that the MDD-associated transcriptome changes derived primarily from myeloid lineage immune cells (classical and non-classical monocytes). A more modest MDD-associated upregulation of glucocorticoid receptor (GR) pathway activity was also present. CMV infection/activity across the combined MDD and HC groups was weakly related to GR pathway activation but not to IRF1 and NF-κB activity; the most salient signature of CMV was activation and/or expansion of the CD8+ T-cell population. The elevated MDD-associated NF-κB (but not IRF1) activity was markedly attenuated after controlling for CMV antibody titer or for CD8+ T-cell prevalence. At least some of the NF-κB signal in MDD may be attributable to the cellular immune response to CMV, suggesting that CMV infection may be one of several pathways contributing to inflammation in depression. The pronounced activation of the antiviral IRF-1 pathway in MDD suggests the contribution of viral processes although this specific antiviral effect was not specific to CMV.CMV may indirectly drive interferon responses by impairing T-cell control of other viral infections.
Collapse
Affiliation(s)
- Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa OK, USA; Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa OK, USA.
| | - Brett A McKinney
- Department of Mathematics and Computer Science, The University of Tulsa, Tulsa, OK, USA
| | - Timothy B Meier
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI USA
| | - Haixia Zheng
- Laureate Institute for Brain Research, Tulsa OK, USA; Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa OK, USA
| | - Bart N Ford
- Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - Robert H Yolken
- Stanley Division of Developmental Neurovirology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - T Kent Teague
- Department of Surgery, University of Oklahoma School of Community Medicine, Tulsa, OK, USA; Department of Psychiatry, University of Oklahoma School of Community Medicine, Tulsa, OK, USA; Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | - Steve W Cole
- University of California, Los Angeles, Cousins Center for Psychoneuroimmunology, Los Angeles, CA, USA; University of California, Los Angeles, David Geffen School of Medicine, Department of Psychiatry and Biobehavioral Sciences, Los Angeles, CA, USA
| |
Collapse
|
3
|
Basova LV, Riley T, Franklin D, Delorme-Walker V, Lim WL, Grant I, Letendre SL, Iudicello JE, Cherner M, Ellis RJ, Marcondes MCG. Identifying methamphetamine use predictors in HIV infection: Immune-dopaminergic signatures in peripheral leukocytes and the role of COMT genotype. Brain Behav Immun Health 2024; 42:100873. [PMID: 39430881 PMCID: PMC11490913 DOI: 10.1016/j.bbih.2024.100873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/24/2024] [Accepted: 09/28/2024] [Indexed: 10/22/2024] Open
Abstract
The pursuit of translational biomarkers is complex due to the heterogeneous human pathophysiology, but critical for disease diagnosis, prognosis, monitoring therapeutic efficacy, and for patient stratification. In HIV-associated neurocognitive impairment (NCI), biomarkers that delineate the trajectory of neuropathogenesis and neurocognitive sequelae are critical, particularly considering confounders such as substance use, including Methamphetamine (METH). METH use is a significant health concern among persons living with HIV (PWH), aggravating cognitive deficits and neuroinflammation despite of antiretrovirals, introducing elements in the microenvironment that are fundamentally differerent in relation to non-METH users, such as high levels of dopamine (DA) affecting HIV-innate immune targets. Yet, current biomarkers do not detect these differences. We hypothesized that predefined DA-induced signatures detectable in peripheral blood leukocytes, can distinguish HIV+ METH users compared to HIV-negative or PWH that are non METH users. The elevated expression of CD8A, CREBBP, CCL5, and combinations of dopaminergic pathway transcripts clustered METH users with detectable CSF viral load and major depressive disorder (MDD), indicating neuroimmune-mechanistic links. Cathecol-o-methyltransferase (COMT) gene polymorphisms affecting DA metabolism improved the identification of PWH using METH with biomarkers. The results indicate that underlying immunedopaminergic mechanisms provide signatures and genotypes that can identify PWH that are METH users and their attributes.
Collapse
Affiliation(s)
- Liana V. Basova
- San Diego Biomedical Research Institute, San Diego, CA, 92121, USA
| | - Tera Riley
- San Diego Biomedical Research Institute, San Diego, CA, 92121, USA
- National Institute for Drug Abuse, Summer Internship, 2023, USA
| | - Donald Franklin
- University of California San Diego, HIV Neurobehavioral Research Program, San Diego, CA, 92103, USA
| | | | - Wei Ling Lim
- San Diego Biomedical Research Institute, San Diego, CA, 92121, USA
| | - Igor Grant
- University of California San Diego, HIV Neurobehavioral Research Program, San Diego, CA, 92103, USA
| | - Scott L. Letendre
- University of California San Diego, HIV Neurobehavioral Research Program, San Diego, CA, 92103, USA
| | - Jennifer E. Iudicello
- University of California San Diego, HIV Neurobehavioral Research Program, San Diego, CA, 92103, USA
| | - Mariana Cherner
- University of California San Diego, HIV Neurobehavioral Research Program, San Diego, CA, 92103, USA
| | - Ronald J. Ellis
- University of California San Diego, HIV Neurobehavioral Research Program, San Diego, CA, 92103, USA
| | | |
Collapse
|
4
|
Ye ML, Wang JK. Association of total bilirubin with depression risk in adults with diabetes: A cross-sectional study. World J Clin Cases 2024; 12:3428-3437. [PMID: 38983435 PMCID: PMC11229937 DOI: 10.12998/wjcc.v12.i18.3428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/30/2024] [Accepted: 05/17/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Individuals with diabetes mellitus are more likely to experience depression, although most patients remain undiagnosed. The relation between total bilirubin and depression has been increasingly discussed, but limited studies have examined the association of total bilirubin with depression risk in adults with diabetes, which warrants attention. AIM To investigate the association between total bilirubin levels and the risk of depression in adults with diabetes. METHODS The study included adults with diabetes from the National Health and Nutrition Examination Survey 2007-2018. Depression was determined using the Patient Health Questionnaire-9. Multivariable logistic regression, propensity score-matched analysis and restricted cubic spline models were utilized to investigate the association between total bilirubin levels and depression risk in adults with diabetes. RESULTS The study included 4758 adults with diabetes, of whom 602 (12.7%) were diagnosed with depression. After adjusting for covariates, we found that diabetic adults with lower total bilirubin levels had a higher risk of depression (OR = 1.230, 95%CI: 1.006-1.503, P = 0.043). This association was further confirmed after propensity score matching (OR = 1.303, 95%CI: 1.034-1.641, P = 0.025). Subgroup analyses showed no significant dependence of age, body mass index, sex, race or hypertension on this association. Restricted cubic spline models displayed an inverted U-shaped association of total bilirubin levels with depression risk within the lower range of total bilirubin levels. The depression risk heightened with the increasing levels of total bilirubin, reaching the highest risk at 6.81 μmol/L and decreasing thereafter. CONCLUSION In adults with diabetes, those with lower levels of total bilirubin were more likely to have depressive symptoms. Serum total bilirubin levels may be used as an additional indicator to assess depression risk in adults with diabetes.
Collapse
Affiliation(s)
- Man-Li Ye
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Jie-Ke Wang
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| |
Collapse
|
5
|
Yu Z, Cole S, Ross K, Hart M, Anis L, Letourneau N. Childhood Adversities and the ATTACH TM Program's Influence on Immune Cell Gene Expression. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2024; 21:776. [PMID: 38929022 PMCID: PMC11204101 DOI: 10.3390/ijerph21060776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/25/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE To determine whether maternal Adverse Childhood Experiences (ACEs) are (a) associated with increased inflammatory gene expression in mother-child dyads and (b) whether a parenting intervention (ATTACH™) moderates the association between maternal ACEs and mother and/or child inflammatory gene expression. METHODS Twenty mother-child dyads, recruited from a domestic violence shelter in Calgary, AB, Canada, were randomized into an ATTACH™ parenting intervention group (n = 9) or a wait-list control group (n = 11). Maternal ACEs were assessed. The mothers and children each provided one non-fasting blood sample after the intervention group completed the ATTACH™ program, which was assayed to quantify the Conserved Transcriptional Response to Adversity (CTRA) score, indicating inflammatory gene expression profile. Mixed-effect linear models were used, separately in mothers and children, to examine the associations between CTRA score, maternal ACEs, and the ACEs-by-intervention group interaction term. The covariates were age, sex, ethnicity, and maternal medication use. RESULTS Higher maternal ACEs were associated with higher child CTRA scores (b = 0.123 ± SE 0.044, p = 0.005), indicating an increased pro-inflammatory gene expression profile. The ATTACH™ parenting intervention moderated this association between maternal ACEs and child CTRA scores (b = 0.328 ± SE 0.133, p = 0.014). In mothers, the ACEs-by-intervention interaction terms were insignificant (p = 0.305). CONCLUSIONS Maternal ACEs could exert an intergenerational impact on child inflammatory activity, and this association could be moderated by participating in the ATTACH™ parenting intervention.
Collapse
Affiliation(s)
- Zhiyuan Yu
- School of Nursing, Johns Hopkins University, 525 N Wolfe St., Baltimore, MD 21205, USA;
| | - Steve Cole
- School of Medicine, University of California-Los Angeles, Le Conte Ave, Los Angeles, CA 10833, USA;
| | - Kharah Ross
- Department of Psychology, Athabasca University, 1 University Dr., Athabasca, AB T9S 3A3, Canada;
| | - Martha Hart
- Alberta Children’s Hospital Research Institute, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada; (M.H.); (L.A.)
| | - Lubna Anis
- Alberta Children’s Hospital Research Institute, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada; (M.H.); (L.A.)
| | - Nicole Letourneau
- Faculty of Nursing & Cumming School of Medicine, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
6
|
Guo Z, Xie Z, Wang P, Li S, Xin X, Wang X. The moderating effect of physical activity on the relationship between neutrophil count and depressive symptoms. Sci Rep 2024; 14:12647. [PMID: 38825659 PMCID: PMC11144697 DOI: 10.1038/s41598-024-63432-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/29/2024] [Indexed: 06/04/2024] Open
Abstract
Variations in immune cell counts can trigger depressive symptoms, while physical activity effectively reduces the risk and severity of depressive symptoms. This study, based on the NHANES database, analyzes the relationship between neutrophil count and depressive symptoms and explores the moderating effect of physical activity on this relationship. Cross-sectional data from the NHANES database were extracted, including immune cell counts, PHQ-9 scores for self-assessment of depressive symptoms, and Global Physical Activity Questionnaire (GPAQ) scores (PA). The interrelations among physical activity, neutrophil count, and depressive symptoms were analyzed. After controlling for confounding factors, neutrophil count was found to have a significant role in identifying depressive symptoms with an odds ratio (OR) [95% Confidence Interval (CI)] = 1.13 [1.02, 1.251]; the moderating effect of physical activity on the impact of neutrophil count on depressive symptoms was statistically significant (coefficient = -0.0028, P < 0.05). Neutrophil count may be a significant factor in identifying depressive symptoms in adults. As an effective moderating factor, physical activity can mitigate the impact of neutrophil count on depressive symptoms to a certain extent.
Collapse
Affiliation(s)
- Zhaohui Guo
- Shanghai University of Sport, Shanghai, 200438, China
| | - Zhenwen Xie
- Shanghai University of Sport, Shanghai, 200438, China
| | - Peng Wang
- Shanghai University of Sport, Shanghai, 200438, China
| | - Shufan Li
- Shanghai University of Sport, Shanghai, 200438, China
| | - Xin Xin
- Shanghai University of Sport, Shanghai, 200438, China
| | - Xing Wang
- Shanghai University of Sport, Shanghai, 200438, China.
| |
Collapse
|
7
|
Gonzales EL, Jeon SJ, Han KM, Yang SJ, Kim Y, Remonde CG, Ahn TJ, Ham BJ, Shin CY. Correlation between immune-related genes and depression-like features in an animal model and in humans. Brain Behav Immun 2023; 113:29-43. [PMID: 37379963 DOI: 10.1016/j.bbi.2023.06.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 06/01/2023] [Accepted: 06/22/2023] [Indexed: 06/30/2023] Open
Abstract
A growing body of evidence suggests that immune-related genes play pivotal roles in the pathophysiology of depression. In the present study, we investigated a plausible connection between gene expression, DNA methylation, and brain structural changes in the pathophysiology of depression using a combined approach of murine and human studies. We ranked the immobility behaviors of 30 outbred Crl:CD1 (ICR) mice in the forced swim test (FST) and harvested their prefrontal cortices for RNA sequencing. Of the 24,532 analyzed genes, 141 showed significant correlations with FST immobility time, as determined through linear regression analysis with p ≤ 0.01. The identified genes were mostly involved in immune responses, especially interferon signaling pathways. Moreover, induction of virus-like neuroinflammation in the brains of two separate mouse cohorts (n = 30 each) using intracerebroventricular polyinosinic:polycytidylic acid injection resulted in increased immobility during FST and similar expression of top immobility-correlated genes. In human blood samples, candidate gene (top 5%) expression profiling using DNA methylation analysis found the interferon-related USP18 (cg25484698, p = 7.04 × 10-11, Δβ = 1.57 × 10-2; cg02518889, p = 2.92 × 10-3, Δβ = - 8.20 × 10-3) and IFI44 (cg07107453, p = 3.76 × 10-3, Δβ = - 4.94 × 10-3) genes to be differentially methylated between patients with major depressive disorder (n = 350) and healthy controls (n = 161). Furthermore, cortical thickness analyses using T1-weighted images revealed that the DNA methylation scores for USP18 were negatively correlated with the thicknesses of several cortical regions, including the prefrontal cortex. Our results reveal the important role of the interferon pathway in depression and suggest USP18 as a potential candidate target. The results of the correlation analysis between transcriptomic data and animal behavior carried out in this study provide insights that could enhance our understanding of depression in humans.
Collapse
Affiliation(s)
- Edson Luck Gonzales
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Se Jin Jeon
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea; Department of Integrative Biotechnology, College of Science and Technology, Sahmyook University, Seoul 01795, Republic of Korea
| | - Kyu-Man Han
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Seung Jin Yang
- Department of Life Science, Handong Global University, Pohang 37554, Republic of Korea
| | - Yujeong Kim
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Chilly Gay Remonde
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea
| | - Tae Jin Ahn
- Department of Life Science, Handong Global University, Pohang 37554, Republic of Korea.
| | - Byung-Joo Ham
- Department of Psychiatry, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea.
| | - Chan Young Shin
- School of Medicine and Center for Neuroscience Research, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
8
|
Rajkumar RP. Examining the Relationships between the Incidence of Infectious Diseases and Mood Disorders: An Analysis of Data from the Global Burden of Disease Studies, 1990-2019. Diseases 2023; 11:116. [PMID: 37754312 PMCID: PMC10528187 DOI: 10.3390/diseases11030116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/27/2023] [Accepted: 09/04/2023] [Indexed: 09/28/2023] Open
Abstract
Mood disorders are among the commonest mental disorders worldwide. Epidemiological and clinical evidence suggests that there are close links between infectious diseases and mood disorders, but the strength and direction of these association remain largely unknown. Theoretical models have attempted to explain this link based on evolutionary or immune-related factors, but these have not been empirically verified. The current study examined cross-sectional and longitudinal associations between the incidence of infectious diseases and mood disorders, while correcting for climate and economic factors, based on data from the Global Burden of Disease Studies, 1990-2019. It was found that major depressive disorder was positively associated with lower respiratory infections, while bipolar disorder was positively associated with upper respiratory infections and negatively associated with enteric and tropical infections, both cross-sectionally and over a period of 30 years. These results suggest that a complex, bidirectional relationship exists between these disorders. This relationship may be mediated through the immune system as well as through the gut-brain and lung-brain axes. Understanding the mechanisms that link these groups of disorders could lead to advances in the prevention and treatment of both.
Collapse
Affiliation(s)
- Ravi Philip Rajkumar
- Department of Psychiatry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry 605006, India
| |
Collapse
|
9
|
Chen H, Wu C, Lv Q, Li M, Ren L. Targeting Mitochondrial Homeostasis: The Role of Acupuncture in Depression Treatment. Neuropsychiatr Dis Treat 2023; 19:1741-1753. [PMID: 37546517 PMCID: PMC10404048 DOI: 10.2147/ndt.s421540] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023] Open
Abstract
Background Depression is a common mental health disorder characterized by persistent feelings of sadness, loss of interest or pleasure, and a range of physical and cognitive symptoms. It affects people of all ages and can significantly impact their daily functioning and quality of life. Mitochondrial homeostasis plays an important role in the pathogenesis of depression. Mitochondrial homeostasis includes mitophagy, mitochondrial oxidative stress, mitoptosis, mitochondrial biogenesis, and mitochondrial dynamics. The regulation of mitochondrial homeostasis is the key link in the prevention and treatment of depression. Methods In this article, we focus on the core link of depression-mitochondrial homeostasis and summarize the research progress of acupuncture targeting mitochondrial homeostasis in the treatment of depression in recent years, so as to provide ideas and experimental basis for the research and formulation of more appropriate depression treatment strategies. Results Acupuncture has been found to regulate mitochondrial homeostasis (by modulating mitochondrial autophagy, reducing mitochondrial oxidative stress, inhibiting mitochondrial fission, inducing mitochondrial biogenesis, and maintaining mitochondrial dynamics), alleviate depression-like behavior, and regulate signal pathways and key proteins. Conclusion Here, we highlight the role of acupuncture in the treatment of depression. A comprehensive exploration of the impact of acupuncture on mitochondrial homeostasis could potentially present a novel mechanism for treating depression and offer fresh perspectives for the treatment of patients with clinical depression.
Collapse
Affiliation(s)
- Haiyang Chen
- Department of Acupuncture and Moxibustion, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Chenlin Wu
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Qin Lv
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Mingjie Li
- Department of Acupuncture and Moxibustion, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Lu Ren
- Graduate School, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
- Mental Disorders Research Laboratory, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| |
Collapse
|
10
|
Sforzini L, Cattaneo A, Ferrari C, Turner L, Mariani N, Enache D, Hastings C, Lombardo G, Nettis MA, Nikkheslat N, Worrell C, Zajkowska Z, Kose M, Cattane N, Lopizzo N, Mazzelli M, Pointon L, Cowen PJ, Cavanagh J, Harrison NA, Jones D, Drevets WC, Mondelli V, Bullmore ET, Pariante CM. Higher immune-related gene expression in major depression is independent of CRP levels: results from the BIODEP study. Transl Psychiatry 2023; 13:185. [PMID: 37264010 PMCID: PMC10235092 DOI: 10.1038/s41398-023-02438-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 06/03/2023] Open
Abstract
Compelling evidence demonstrates that some individuals suffering from major depressive disorder (MDD) exhibit increased levels of inflammation. Most studies focus on inflammation-related proteins, such as serum or plasma C-reactive protein (CRP). However, the immune-related modifications associated with MDD may be not entirely captured by CRP alone. Analysing mRNA gene expression levels, we aimed to identify broader molecular immune-related phenotypes of MDD. We examined 168 individuals from the non-interventional, case-control, BIODEP study, 128 with a diagnosis of MDD and 40 healthy controls. Individuals with MDD were further divided according to serum high-sensitivity (hs)CRP levels (n = 59 with CRP <1, n = 33 with CRP 1-3 and n = 36 with CRP >3 mg/L). We isolated RNA from whole blood and performed gene expression analyses using RT-qPCR. We measured the expression of 16 immune-related candidate genes: A2M, AQP4, CCL2, CXCL12, CRP, FKBP5, IL-1-beta, IL-6, ISG15, MIF, GR, P2RX7, SGK1, STAT1, TNF-alpha and USP18. Nine of the 16 candidate genes were differentially expressed in MDD cases vs. controls, with no differences between CRP-based groups. Only CRP mRNA was clearly associated with serum CRP. In contrast, plasma (proteins) IL-6, IL-7, IL-8, IL-10, IL-12/IL-23p40, IL-16, IL-17A, IFN-gamma and TNF-alpha, and neutrophils counts, were all differentially regulated between CRP-based groups (higher in CRP >3 vs. CRP <1 and/or controls), reflecting the gradient of CRP values. Secondary analyses on MDD individuals and controls with CRP values <1 mg/L (usually interpreted as 'no inflammation') confirmed MDD cases still had significantly different mRNA expression of immune-related genes compared with controls. These findings corroborate an immune-related molecular activation in MDD, which appears to be independent of serum CRP levels. Additional biological mechanisms may then be required to translate this mRNA signature into inflammation at protein and cellular levels. Understanding these mechanisms will help to uncover the true immune abnormalities in depression, opening new paths for diagnosis and treatment.
Collapse
Affiliation(s)
- Luca Sforzini
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RT, UK.
| | - Annamaria Cattaneo
- Biological Psychiatric Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Clarissa Ferrari
- Research and Clinical Trials Service, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, 25124, Italy
| | - Lorinda Turner
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Nicole Mariani
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RT, UK
| | - Daniela Enache
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RT, UK
| | - Caitlin Hastings
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RT, UK
| | - Giulia Lombardo
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RT, UK
| | - Maria A Nettis
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RT, UK
| | - Naghmeh Nikkheslat
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RT, UK
| | - Courtney Worrell
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RT, UK
| | - Zuzanna Zajkowska
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RT, UK
| | - Melisa Kose
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RT, 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
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Monica Mazzelli
- Biological Psychiatric Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125, Brescia, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, 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, School of Infection & Immunity, University of Glasgow, G12 8TA, Glasgow, Scotland
| | - Neil A Harrison
- School of Medicine, School of Psychology, Cardiff University Brain Research Imaging Centre, Maindy Road, Cardiff, CF24 4HQ, UK
| | - 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
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RT, UK
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| | - Edward T Bullmore
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Carmine M Pariante
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Psychological Medicine, Maurice Wohl Clinical Neuroscience Institute, London, SE5 9RT, UK
- National Institute for Health Research (NIHR) Mental Health Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| |
Collapse
|
11
|
Sani G, Margoni S, Brugnami A, Ferrara OM, Bernardi E, Simonetti A, Monti L, Mazza M, Janiri D, Moccia L, Kotzalidis GD, Chieffo DPR, Janiri L. The Nrf2 Pathway in Depressive Disorders: A Systematic Review of Animal and Human Studies. Antioxidants (Basel) 2023; 12:antiox12040817. [PMID: 37107192 PMCID: PMC10135298 DOI: 10.3390/antiox12040817] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
There is increasing interest in the involvement of antioxidative systems in protecting from depression. Among these, Nrf2 occupies a central place. We aimed to review the role of Nrf2 in depression. For this reason, we conducted a PubMed search using as search strategy (psychiatr*[ti] OR schizo*[ti] OR psychot*[ti] OR psychos*[ti] OR depress*[ti] OR MDD[ti] OR BD[ti] OR bipolar[ti] OR Anxiety[ti] OR antidepress*[ti] OR panic[ti] OR obsess*[ti] OR compulsio*[ti] OR “mood disord*”[ti] OR phobi*[ti] OR agoraphob*[ti] OR anorex*[ti] OR anorect*[ti] OR bulimi*[ti] OR “eating disorder*”[ti] OR neurodevelopm*[ti] OR retardation[ti] OR autism[ti] OR autistic[ti] OR ASM[ti] OR adhd[ti] OR “attention-deficit”[ti]) AND nrf2, which on the 9th of March produced 208 results of which 89 were eligible for our purposes. Eligible articles were studies reporting data of Nrf2 manipulations or content by any treatment in human patients or animals with any animal model of depression. Most studies were on mice only (N = 58), 20 on rats only, and three on both rats and mice. There were two studies on cell lines (in vitro) and one each on nematodes and fish. Only four studies were conducted in humans, one of which was post mortem. Most studies were conducted on male animals; however, human studies were carried out on both men and women. The results indicate that Nrf2 is lower in depression and that antidepressant methods (drugs or other methods) increase it. Antioxidant systems and plasticity-promoting molecules, such as those in the Nrf2–HO-1, BDNF–TrkB, and cyclic AMP–CREB pathways, could protect from depression, while glycogen synthase kinase-3β and nuclear factor κB oppose these actions, thus increasing depressive-like behaviours. Since Nrf2 is also endowed with tumorigenic and atherogenic potential, the balance between benefits and harms must be taken into account in designing novel drugs aiming at increasing the intracellular content of Nrf2.
Collapse
Affiliation(s)
- Gabriele Sani
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Department of Neuroscience, Head, Neck and Thorax, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
- Correspondence:
| | - Stella Margoni
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Andrea Brugnami
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Ottavia Marianna Ferrara
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Evelina Bernardi
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Alessio Simonetti
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Department of Neuroscience, Head, Neck and Thorax, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
- Centro Lucio Bini, Via Crescenzio 42, 00193 Rome, Italy
| | - Laura Monti
- UOS Clinical Psychology, Clinical Government, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| | - Marianna Mazza
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Department of Neuroscience, Head, Neck and Thorax, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| | - Delfina Janiri
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Department of Neuroscience, Head, Neck and Thorax, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| | - Lorenzo Moccia
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Department of Neuroscience, Head, Neck and Thorax, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| | - Georgios D. Kotzalidis
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- NESMOS Department, Faculty of Medicine and Psychology, Sant’Andrea University Hospital, University of Rome La Sapienza, Via di Grottarossa, 1035-1039, 00189 Rome, Italy
| | - Daniela Pia Rosaria Chieffo
- UOS Clinical Psychology, Clinical Government, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| | - Luigi Janiri
- Institute of Psychiatry, Department of Neuroscience, Catholic University of the Sacred Hearth, Rome, Largo Francesco Vito 1, 00168 Rome, Italy
- Department of Psychiatry, Department of Neuroscience, Head, Neck and Thorax, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 1, 00168 Rome, Italy
| |
Collapse
|
12
|
The genomic impact of kindness to self vs. others: A randomized controlled trial. Brain Behav Immun 2022; 106:40-48. [PMID: 35905861 DOI: 10.1016/j.bbi.2022.07.159] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE Prosocial behavior has been linked to improved physical health, but the biological mechanisms involved remain unclear. This study tested whether a 4-week kindness intervention could reduce expression of a stress-related immune response gene signature known as the Conserved Transcriptional Response to Adversity (CTRA). METHODS In a diverse sample of community adults (N = 182), study participants were randomly assigned to perform 3 kind acts for other people, to perform 3 kind acts for themselves, or to list daily activities (control), on one day per week over 4 weeks. CTRA gene expression was measured by RNA sequencing of dried blood spots (DBS) collected at baseline and 5 weeks later (1 week after completing study assignments). Participants' descriptions of their kind acts were coded for protocol adherence and act content. RESULTS Participants who were randomized to perform kind acts for others showed significant reductions in CTRA gene expression relative to controls. Participants who were randomized to perform kind acts for themselves also showed significant reductions in CTRA gene expression relative to controls, but this pattern emerged only for those who failed to perform the requested self-kind acts (protocol non-adherent). Those who fully adhered to the self-kindness protocol showed no change in CTRA gene expression and did not differ from controls. Act content analyses implicated self-stress-reducing behavior in the paradoxical effects of self-kindness and the physical presence of others in the effects of prosocial behavior. CONCLUSIONS Prosocial engagement-doing something kind for others rather than oneself-reduces CTRA gene expression. The nature of kind acts and their intended recipient plays a key role in shaping the genomic impact of kindness.
Collapse
|
13
|
The Problem of Malnutrition Associated with Major Depressive Disorder from a Sex-Gender Perspective. Nutrients 2022; 14:nu14051107. [PMID: 35268082 PMCID: PMC8912662 DOI: 10.3390/nu14051107] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 02/03/2023] Open
Abstract
Major depressive disorder (MDD) is an incapacitating condition characterized by loss of interest, anhedonia and low mood, which affects almost 4% of people worldwide. With rising prevalence, it is considered a public health issue that affects economic productivity and heavily increases health costs alone or as a comorbidity for other pandemic non-communicable diseases (such as obesity, cardiovascular disease, diabetes, inflammatory bowel diseases, etc.). What is even more noteworthy is the double number of women suffering from MDD compared to men. In fact, this sex-related ratio has been contemplated since men and women have different sexual hormone oscillations, where women meet significant changes depending on the age range and moment of life (menstruation, premenstruation, pregnancy, postpartum, menopause…), which seem to be associated with susceptibility to depressive symptoms. For instance, a decreased estrogen level promotes decreased activation of serotonin transporters. Nevertheless, sexual hormones are not the only triggers that alter neurotransmission of monoamines and other neuropeptides. Actually, different dietary habits and/or nutritional requirements for specific moments of life severely affect MDD pathophysiology in women. In this context, the present review aims to descriptively collect information regarding the role of malnutrition in MDD onset and course, focusing on female patient and especially macro- and micronutrient deficiencies (amino acids, ω3 polyunsaturated fatty acids (ω3 PUFAs), folate, vitamin B12, vitamin D, minerals…), besides providing evidence for future nutritional intervention programs with a sex-gender perspective that hopefully improves mental health and quality of life in women.
Collapse
|
14
|
Behl T, Rana T, Alotaibi GH, Shamsuzzaman M, Naqvi M, Sehgal A, Singh S, Sharma N, Almoshari Y, Abdellatif AAH, Iqbal MS, Bhatia S, Al-Harrasi A, Bungau S. Polyphenols inhibiting MAPK signalling pathway mediated oxidative stress and inflammation in depression. Biomed Pharmacother 2021; 146:112545. [PMID: 34922112 DOI: 10.1016/j.biopha.2021.112545] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 12/24/2022] Open
Abstract
Depression is one of the most debilitating psychiatric disorders affecting people of all ages worldwide. Despite significant heterogeneity between studies, increased inflammation and oxidative stress have been found in depression. Oxidative stress and inflammation are involved in the pathogenesis of depression. In the current review, we discussed the markers of oxidative stress and inflammation in depressive disorder and the association between these markers and the antidepressant treatment. The role of natural polyphenols in regulating various cell signaling pathways related to oxidative stress and inflammation has also been reviewed. The inhibitory effect of polyphenols on several cell signaling pathways reveals the vital role of polyphenols in the prevention and treatment of depressive disorder. Understanding the mechanism of polyphenols implicated in the regulation of cell signaling pathways is essential for the identification of lead compounds and the development of novel effective compounds for the prevention and treatment of depressive disorder.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Tarapati Rana
- Chitkara College of Pharmacy, Chitkara University, Punjab, India; Government Pharmacy College, Seraj, Mandi, Himachal Pradesh, India
| | - Ghallab H Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Kingdom of Saudi Arabia
| | - Md Shamsuzzaman
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Kingdom of Saudi Arabia
| | - Maaz Naqvi
- Central Research Laboratory, Department of Pharmacology, HIMSR, Jamia Hamdard, India
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Yosif Almoshari
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Muhammad Shahid Iqbal
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman; School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Nizwa, Oman
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania.
| |
Collapse
|
15
|
Transcriptomic signatures of psychomotor slowing in peripheral blood of depressed patients: evidence for immunometabolic reprogramming. Mol Psychiatry 2021; 26:7384-7392. [PMID: 34535767 PMCID: PMC8881295 DOI: 10.1038/s41380-021-01258-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/25/2021] [Accepted: 07/30/2021] [Indexed: 02/08/2023]
Abstract
Inflammation impacts basal ganglia motor circuitry in association with psychomotor retardation, a key symptom of major depression (MD). We previously reported associations between circulating protein inflammatory biomarkers and psychomotor slowing as measured by neuropsychological tests probing psychomotor speed in patients with MD. To discover novel transcriptional signatures in peripheral blood immune cells related to psychomotor slowing, microarray data were analyzed in a primary cohort of 88 medically-stable, unmedicated, ambulatory MD patients. Results were confirmed and extended in a second cohort of 57 patients with treatment resistant depression (TRD) before and after anti-inflammatory challenge with the tumor necrosis factor antagonist infliximab versus placebo. Composite scores reflecting pure motor and cognitive-motor processing speed were linearly associated with 403 and 266 gene transcripts in each cohort, respectively (|R| > 0.30, p < 0.01), that were enriched for cytokine signaling and glycolysis-related pathways (p < 0.05). Unsupervised clustering in the primary cohort revealed two psychomotor slowing-associated gene co-expression modules that were enriched for interferon, interleukin-6, aerobic glycolysis, and oxidative phosphorylation pathways (p < 0.05, q < 0.1). Transcripts were predominantly derived from monocytes, plasmacytoid dendritic cells, and natural killer cells (p's < 0.05). In infliximab-treated TRD patients with high plasma C-reactive protein concentrations (>5 mg/L), two differential co-expression modules enriched for oxidative stress and mitochondrial degradation were associated with improvements in psychomotor reaction time (p < 0.05). These results indicate that inflammatory signaling and associated metabolic reprogramming in peripheral blood immune cells are associated with systemic inflammation in depression and may affect relevant brain circuits to promote psychomotor slowing.
Collapse
|
16
|
Moisan MP, Foury A, Dexpert S, Cole SW, Beau C, Forestier D, Ledaguenel P, Magne E, Capuron L. Transcriptomic signaling pathways involved in a naturalistic model of inflammation-related depression and its remission. Transl Psychiatry 2021; 11:203. [PMID: 33824279 PMCID: PMC8024399 DOI: 10.1038/s41398-021-01323-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/19/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023] Open
Abstract
This study aimed at identifying molecular biomarkers of inflammation-related depression in order to improve diagnosis and treatment. For this, we performed whole-genome expression profiling from peripheral blood in a naturalistic model of inflammation-associated major depressive disorder (MDD) represented by comorbid depression in obese patients. We took advantage of the marked reduction of depressive symptoms and inflammation following bariatric surgery to test the robustness of the identified biomarkers. Depression was assessed during a clinical interview using Mini-International Neuropsychiatric Interview and the 10-item, clinician-administered, Montgomery-Asberg Depression Rating Scale. From a cohort of 100 massively obese patients, we selected 33 of them for transcriptomic analysis. Twenty-four of them were again analyzed 4-12 months after bariatric surgery. We conducted differential gene expression analyses before and after surgery in unmedicated MDD and non-depressed obese subjects. We found that TP53 (Tumor Protein 53), GR (Glucocorticoid Receptor), and NFκB (Nuclear Factor kappa B) pathways were the most discriminating pathways associated with inflammation-related MDD. These signaling pathways were processed in composite z-scores of gene expression that were used as biomarkers in regression analyses. Results showed that these transcriptomic biomarkers highly predicted depressive symptom intensity at baseline and their remission after bariatric surgery. While inflammation was present in all patients, GR signaling over-activation was found only in depressed ones where it may further increase inflammatory and apoptosis pathways. In conclusion, using an original model of inflammation-related depression and its remission without antidepressants, we provide molecular predictors of inflammation-related MDD and new insights in the molecular pathways involved.
Collapse
Affiliation(s)
- Marie-Pierre Moisan
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France.
| | - Aline Foury
- grid.488493.a0000 0004 0383 684XUniv. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Sandra Dexpert
- grid.488493.a0000 0004 0383 684XUniv. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Steve W. Cole
- grid.19006.3e0000 0000 9632 6718Division of Hematology-Oncology, Department of Psychiatry & Biobehavioral Sciences and Department of Medicine, UCLA School of Medicine, Los Angeles, CA USA
| | - Cédric Beau
- Service de Chirurgie Digestive et Pariétale, Clinique Tivoli, Bordeaux, and Clinique Jean Villar, Bruges, France
| | - Damien Forestier
- Service de Chirurgie Digestive et Pariétale, Clinique Tivoli, Bordeaux, and Clinique Jean Villar, Bruges, France
| | - Patrick Ledaguenel
- Service de Chirurgie Digestive et Pariétale, Clinique Tivoli, Bordeaux, and Clinique Jean Villar, Bruges, France
| | - Eric Magne
- Service de Chirurgie Digestive et Pariétale, Clinique Tivoli, Bordeaux, and Clinique Jean Villar, Bruges, France
| | - Lucile Capuron
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France.
| |
Collapse
|
17
|
Nobis A, Zalewski D, Waszkiewicz N. Peripheral Markers of Depression. J Clin Med 2020; 9:E3793. [PMID: 33255237 PMCID: PMC7760788 DOI: 10.3390/jcm9123793] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 12/22/2022] Open
Abstract
Major Depressive Disorder (MDD) is a leading cause of disability worldwide, creating a high medical and socioeconomic burden. There is a growing interest in the biological underpinnings of depression, which are reflected by altered levels of biological markers. Among others, enhanced inflammation has been reported in MDD, as reflected by increased concentrations of inflammatory markers-C-reactive protein, interleukin-6, tumor necrosis factor-α and soluble interleukin-2 receptor. Oxidative and nitrosative stress also plays a role in the pathophysiology of MDD. Notably, increased levels of lipid peroxidation markers are characteristic of MDD. Dysregulation of the stress axis, along with increased cortisol levels, have also been reported in MDD. Alterations in growth factors, with a significant decrease in brain-derived neurotrophic factor and an increase in fibroblast growth factor-2 and insulin-like growth factor-1 concentrations have also been found in MDD. Finally, kynurenine metabolites, increased glutamate and decreased total cholesterol also hold promise as reliable biomarkers for MDD. Research in the field of MDD biomarkers is hindered by insufficient understanding of MDD etiopathogenesis, substantial heterogeneity of the disorder, common co-morbidities and low specificity of biomarkers. The construction of biomarker panels and their evaluation with use of new technologies may have the potential to overcome the above mentioned obstacles.
Collapse
Affiliation(s)
- Aleksander Nobis
- Department of Psychiatry, Medical University of Bialystok, pl. Brodowicza 1, 16-070 Choroszcz, Poland; (D.Z.); (N.W.)
| | | | | |
Collapse
|
18
|
Goldsmith DR, Bekhbat M, Le NA, Chen X, Woolwine BJ, Li Z, Haroon E, Felger JC. Protein and gene markers of metabolic dysfunction and inflammation together associate with functional connectivity in reward and motor circuits in depression. Brain Behav Immun 2020; 88:193-202. [PMID: 32387344 PMCID: PMC7415617 DOI: 10.1016/j.bbi.2020.05.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/04/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022] Open
Abstract
Bidirectional relationships between inflammation and metabolic dysfunction may contribute to the pathophysiology of psychiatric illnesses like depression. Metabolic disturbances drive inflammation, which in turn exacerbate metabolic outcomes including insulin resistance. Both inflammatory (e.g. endotoxin, vaccination) and metabolic challenges (e.g. glucose ingestion) have been shown to affect activity and functional connectivity (FC) in brain regions that subserve reward and motor processing. We previously reported relationships between elevated concentrations of endogenous inflammatory markers including C-reactive protein (CRP) and low corticostriatal FC, which correlated with symptoms of anhedonia and motor slowing in major depression (MD). Herein, we examined whether similar relationships were observed between plasma markers related to glucose metabolism (non-fasting concentrations of glucose, insulin, leptin, adiponectin and resistin) in 42 medically-stable, unmedicated MD outpatients who underwent fMRI. A targeted, hypothesis-driven approach was used to assess FC between seeds in subdivisions of the ventral and dorsal striatum and a region in ventromedial prefrontal cortex (VS-vmPFC), which was previously found to correlate with both inflammation and symptoms of anhedonia and motor slowing. Associations between FC and gene expression signatures were also explored. A composite score of all 5 glucose-related markers (with increasing values reflecting higher concentrations) was negatively correlated with both ventral striatum (VS)-vmPFC (r = -0.33, p < 0.05) and dorsal caudal putamen (dcP)-vmPFC (r = -0.51, p < 0.01) FC, and remained significant after adjusting for covariates including body mass index (p < 0.05). Moreover, an interaction between the glucose-related composite score and CRP was observed for these relationships (F[2,33] = 4.3, p < 0.05) whereby significant correlations between the glucose-related metabolic markers and FC was found only in patients with high plasma CRP (>3 mg/L; r = -0.61 to -0.81, p < 0.05). Insulin and resistin were the individual markers most predictive of VS-vmPFC and dcP-mPFC FC, respectively, and insulin, resistin and CRP clustered together and in association with both LV-vmPFC and dcP-vmPFC in principal component analyses. Exploratory whole blood gene expression analyses also confirmed that gene probes negatively associated with FC were enriched for both inflammatory and metabolic pathways (FDR p < 0.05). These results provide preliminary evidence that inflammation and metabolic dysfunction contribute jointly to deficits in reward and motor circuits in MD. Future studies using fasting samples and longitudinal and interventional approaches are required to further elucidate the respective contributions of inflammation and metabolic dysfunction to circuits and symptoms relevant to motivation and motor activity, which may have treatment implications for patients with psychiatric illnesses like depression.
Collapse
Affiliation(s)
- David R Goldsmith
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, United States
| | - Mandakh Bekhbat
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, United States
| | - Ngoc-Anh Le
- Biomarker Core Laboratory, Foundation for Atlanta Veterans Education and Research, Atlanta VAHSC, Decatur, GA 30033, United States
| | - Xiangchuan Chen
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, United States
| | - Bobbi J Woolwine
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, United States
| | - Zhihao Li
- School of Psychology, Shenzhen University, Shenzhen, Guangdong 518060, China; Center for Brain Disorders and Cognitive Neuroscience, Shenzhen University, Shenzhen, Guangdong 518060, China.
| | - Ebrahim Haroon
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, United States; The Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States.
| | - Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, United States; The Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States.
| |
Collapse
|
19
|
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: 62] [Impact Index Per Article: 12.4] [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
|
20
|
Bhatt S, Nagappa AN, Patil CR. Role of oxidative stress in depression. Drug Discov Today 2020; 25:1270-1276. [DOI: 10.1016/j.drudis.2020.05.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 04/25/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022]
|
21
|
A genetic profile of refractory individuals with major depressive disorder and their responsiveness to transcranial magnetic stimulation. Brain Stimul 2020; 13:1091-1093. [PMID: 32387243 DOI: 10.1016/j.brs.2020.04.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/28/2020] [Indexed: 01/13/2023] Open
|
22
|
Visentin APV, Colombo R, Scotton E, Fracasso DS, da Rosa AR, Branco CS, Salvador M. Targeting Inflammatory-Mitochondrial Response in Major Depression: Current Evidence and Further Challenges. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2972968. [PMID: 32351669 PMCID: PMC7178465 DOI: 10.1155/2020/2972968] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/26/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
The prevalence of psychiatric disorders has increased in recent years. Among existing mental disorders, major depressive disorder (MDD) has emerged as one of the leading causes of disability worldwide, affecting individuals throughout their lives. Currently, MDD affects 15% of adults in the Americas. Over the past 50 years, pharmacotherapy, psychotherapy, and brain stimulation have been used to treat MDD. The most common approach is still pharmacotherapy; however, studies show that about 40% of patients are refractory to existing treatments. Although the monoamine hypothesis has been widely accepted as a molecular mechanism to explain the etiology of depression, its relationship with other biochemical phenomena remains only partially understood. This is the case of the link between MDD and inflammation, mitochondrial dysfunction, and oxidative stress. Studies have found that depressive patients usually exhibit altered inflammatory markers, mitochondrial membrane depolarization, oxidized mitochondrial DNA, and thus high levels of both central and peripheral reactive oxygen species (ROS). The effect of antidepressants on these events remains unclear. Nevertheless, the effects of ROS on the brain are well known, including lipid peroxidation of neuronal membranes, accumulation of peroxidation products in neurons, protein and DNA damage, reduced antioxidant defenses, apoptosis induction, and neuroinflammation. Antioxidants such as ascorbic acid, tocopherols, and coenzyme Q have shown promise in some depressive patients, but without consensus on their efficacy. Hence, this paper provides a review of MDD and its association with inflammation, mitochondrial dysfunction, and oxidative stress and is aimed at thoroughly discussing the putative links between these events, which may contribute to the design and development of new therapeutic approaches for patients.
Collapse
Affiliation(s)
| | - Rafael Colombo
- Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, RS 95070 560, Brazil
| | - Ellen Scotton
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Farmacologia e Terapêutica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Débora Soligo Fracasso
- Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, RS 95070 560, Brazil
| | - Adriane Ribeiro da Rosa
- Laboratório de Psiquiatria Molecular, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Catia Santos Branco
- Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, RS 95070 560, Brazil
| | - Mirian Salvador
- Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, RS 95070 560, Brazil
| |
Collapse
|
23
|
Chiang JJ, Cole SW, Bower JE, Irwin MR, Taylor SE, Arevalo J, Fuligni AJ. Depressive symptoms and immune transcriptional profiles in late adolescents. Brain Behav Immun 2019; 80:163-169. [PMID: 30851376 PMCID: PMC6710012 DOI: 10.1016/j.bbi.2019.03.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 12/31/2018] [Accepted: 03/05/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Rates of depression increase and peak during late adolescence and alterations in immune processes are thought to be both a risk factor and outcome of depression. However, few studies have examined depression-immune dynamics among adolescents. Using a functional genomics approach, the current study examined whether depressive symptoms were associated with activation of a gene expression profile, characterized by upregulated expression of pro-inflammatory-related genes and downregulated expression of antiviral-related genes in a sample of older adolescents (Mage = 18.37, SD = 0.51). METHOD Participants (n = 87) reported on their depressive symptoms during the past week using the CES-D, and provided blood samples for genome-wide transcriptional profiling of mRNA. RESULTS Adolescents with clinically-significant levels of depressive symptoms (CES-D ≥ 16) exhibited upregulated expression of inflammation-related genes and downregulated expression of antiviral-related genes compared to their peers with lower levels of depressive symptoms (CES-D < 16). Bioinformatics analyses suggested that this pattern of differential gene expression was mediated by greater activity of the pro-inflammatory transcription factor, nuclear factor-kappa B (NF-κB), and reduced activity of glucocorticoid receptors (GRs) and interferon response factors (IRFs). Additional analyses implicated monocytes, B cells, and dendritic cells as primary cellular sources of the observed gene expression patterns associated with depressive symptoms. CONCLUSION Results are consistent with past work demonstrating links between depression and altered immunity. They provide a molecular basis for these associations and suggest that the underlying molecular signature may emerge as early as late adolescence when rates of depression tend to increase.
Collapse
Affiliation(s)
| | - Steve W. Cole
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles,Cousins Center for Psychoneuroimmunology, University of California, Los Angeles
| | - Julienne E. Bower
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles,Cousins Center for Psychoneuroimmunology, University of California, Los Angeles,Department of Psychology, University of California, Los Angeles
| | - Michael R. Irwin
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles,Cousins Center for Psychoneuroimmunology, University of California, Los Angeles,Department of Psychology, University of California, Los Angeles
| | | | - Jesusa Arevalo
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles,Cousins Center for Psychoneuroimmunology, University of California, Los Angeles
| | - Andrew J. Fuligni
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles,Cousins Center for Psychoneuroimmunology, University of California, Los Angeles,Department of Psychology, University of California, Los Angeles
| |
Collapse
|
24
|
Transcriptomic predictors of inflammation-induced depressed mood. Neuropsychopharmacology 2019; 44:923-929. [PMID: 30643228 PMCID: PMC6462041 DOI: 10.1038/s41386-019-0316-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/11/2018] [Accepted: 01/05/2019] [Indexed: 02/07/2023]
Abstract
Inflammation plays a significant role in the pathophysiology of depression. However, not all individuals exposed to inflammatory challenge develop depression, and identifying those at risk is necessary to develop targeted monitoring, prevention, and treatment strategies. Within a randomized double-blind placebo-controlled study (n = 115), we examined whether leukocyte transcriptome profiles predicted inflammation-induced depressed mood in volunteers who received low-dose intravenous endotoxin (n = 58; aged 18-50). At baseline, transcription factor (TF) activities were assessed using genome-wide transcriptional profiling of peripheral blood mononuclear cells and promoter-based bioinformatic analyses. Then, participants were administered endotoxin. Self-reported depressed mood was assessed using the Profile of Mood States. Based on extant studies linking transcriptional profiles to depressive disorder, we examined whether post-endotoxin depressed mood is predicted by baseline activity of TFs related to immune activation, sympathetic activation, and glucocorticoid insensitivity: respectively, nuclear factor kappa B (NF-kB), cAMP response element-binding protein (CREB), and glucocorticoid receptor (GR). Twenty-one participants (36%) experienced an increase in depressed mood from baseline to 2 h post endotoxin, when depressive response peaks. Bioinformatics analyses controlling for age, sex, ethnicity, body mass index, and physical sickness response revealed that post-endotoxin depressed mood was predicted by increased baseline activity of TFs related to inflammation (NF-kB) and beta-adrenergic signaling (CREB) and by decreased activity of GR-related TFs (P's < 0.001). Inflammation-induced depressed mood is predicted by peripheral transcriptome profiles related to immune activation, sympathetic activation, and glucocorticoid insensitivity. With further replication, these stress-related molecular profiles could be used for a novel genomic approach for identifying individuals at high-risk for the inflammatory subtype of depression.
Collapse
|
25
|
Abstract
Gene expression profiling studies of people exposed to chronic threat have identified a Conserved Transcriptional Response to Adversity (CTRA) in circulating immune cells. This physiological pattern is characterized by up-regulated expression of genes involved in inflammation and down-regulated expression of genes involved in Type I interferon responses. The CTRA is mediated by beta-adrenergic signaling pathways that transduce sympathetic nervous system activity into changes in transcription factor activity and hematopoietic output of myeloid lineage immune cells (monocytes, neutrophils, and dendritic cells). Recent research has begun to identify the CNS processes that regulate peripheral CTRA activity, define its implications for disease, and explore the role of positive psychosocial factors in buffering such effects. The CTRA provides a genomic framework for understanding PNI relationships and connecting macro-level psychosocial processes to the micro-level biology of health and disease.
Collapse
Affiliation(s)
- Steven W Cole
- UCLA School of Medicine, Prepared for Current Opinion in Behavioral Science - Psychoneuroimmunology
| |
Collapse
|
26
|
Zhou Y, Lutz PE, Wang YC, Ragoussis J, Turecki G. Global long non-coding RNA expression in the rostral anterior cingulate cortex of depressed suicides. Transl Psychiatry 2018; 8:224. [PMID: 30337518 PMCID: PMC6193959 DOI: 10.1038/s41398-018-0267-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 07/10/2018] [Accepted: 09/10/2018] [Indexed: 01/17/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are an emerging class of regulatory RNA that may be implicated in psychiatric disorders. Here we performed RNA-sequencing in the rostral anterior cingulate cortex of 26 depressed suicides and 24 matched controls. We first performed differential lncRNA expression analysis, and then conducted Weighted Gene Co-expression Network Analysis (WGCNA) to identify co-expression modules associating with depression and suicide. We identified 23 differentially expressed lncRNAs (FDR < 0.1) as well as their differentially expressed overlapping and antisense protein-coding genes. Several of these overlapping or antisense genes were associated with interferon signaling, which is a component of the innate immune response. Using WGCNA, we identified modules of highly co-expressed genes associated with depression and suicide and found protein-coding genes highly connected to differentially expressed lncRNAs within these modules. These protein-coding genes were located distal to their associated lncRNAs and were found to be part of several GO terms enriched in the significant modules, which include: cytoskeleton organization, plasma membrane, cell adhesion, nucleus, DNA-binding, and regulation of dendrite development and morphology. Altogether, we report that lncRNAs are differentially expressed in the brains of depressed individuals who died by suicide and may represent regulators of important molecular functions and biological processes.
Collapse
Grants
- R01 DA033684 NIDA NIH HHS
- Dr. G. Turecki holds a Canada Research Chair (Tier 1), FRQS Chercheur National salary award and a NARSAD Distinguished Investigator Award; he is supported by grants FDN148374, MOP93775, MOP11260, MOP119429, and MOP119430 from CIHR, by NIH grant 1R01DA033684, by the FRQS through the Quebec Network on Suicide, Mood Disorders, and Related Disorders, and through an investigator-initiated research grant from Pfizer
- Scholarships from the Fondation Fyssen, the Fondation Bettencourt-Schueller, the Canadian Institutes of Health Research, the American Foundation of Suicide Prevention, the Fondation Deniker and the Fondation pour la Recherche Médicale.
- CFI grant number 32557 and Genome Canada Genome Innovation Node awards.
Collapse
Affiliation(s)
- Yi Zhou
- McGill Group for Suicide Studies, McGill University, 6875 LaSalle Boulevard, Montreal, QC, H4H 1R3, Canada
| | - Pierre-Eric Lutz
- McGill Group for Suicide Studies, McGill University, 6875 LaSalle Boulevard, Montreal, QC, H4H 1R3, Canada
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, France
| | - Yu Chang Wang
- McGill University and Génome Québec Innovation Centre, 740 Dr. Penfield Avenue, Room 7104, Montréal, QC, H3A 0G1, Canada
| | - Jiannis Ragoussis
- McGill University and Génome Québec Innovation Centre, 740 Dr. Penfield Avenue, Room 7104, Montréal, QC, H3A 0G1, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, McGill University, 6875 LaSalle Boulevard, Montreal, QC, H4H 1R3, Canada.
| |
Collapse
|
27
|
Hashimoto K. Essential Role of Keap1-Nrf2 Signaling in Mood Disorders: Overview and Future Perspective. Front Pharmacol 2018; 9:1182. [PMID: 30386243 PMCID: PMC6198170 DOI: 10.3389/fphar.2018.01182] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 09/28/2018] [Indexed: 12/11/2022] Open
Abstract
Depression is one of the most common mood disorders with a high rate of relapse. Accumulating evidence suggests that the transcription factor Kelch-like erythroid cell-derived protein with CNC homology (ECH)-associated protein 1 (Keap1)-Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) system plays a key role in inflammation which is involved in depression. Preclinical studies demonstrated that the protein expressions of Keap1 and Nrf2 in the prefrontal cortex (PFC), CA3 and dentate gyrus (DG) of hippocampus in mice with depression-like phenotype were lower than control mice. In the learned helplessness paradigm, the protein levels of Keap1 and Nrf2 in the PFC and DG of hippocampus from rats with depression-like phenotype were also lower than control and resilient rats. Furthermore, rodents with depression-like phenotype have higher levels of pro-inflammatory cytokines. Interestingly, Nrf2 knock-out (KO) mice exhibit depression-like phenotype, and higher serum levels of pro-inflammatory cytokines compared with wild-type mice. Furthermore, Nrf2 KO mice have lower expression of brain-derived neurotrophic factor (BDNF) in the PFC, and CA3 and DG of hippocampus compared to wild-type mice. 7,8-Dihydroxyflavone, a TrkB agonist, showed antidepressant effects in Nrf2 KO mice, by stimulating BDNF-TrkB in the PFC, CA3, and DG. Pretreatment with sulforaphane, a naturally occurring Nrf2 activator, prevented depression-like phenotype in mice after inflammation, or chronic social defeat stress. Interestingly, dietary intake of 0.1% glucoraphanin (a precursor of sulforaphane) containing food during juvenile and adolescent stages of mice could prevent depression-like phenotype in adulthood after chronic social defeat stress. Moreover, the protein expressions of Keap1 and Nrf2 in the parietal cortex from major depressive disorder and bipolar disorder were lower than controls. These findings suggest that Keap1-Nrf2 system plays a key role in the stress resilience which is involved in the pathophysiology of mood disorders. It is, therefore, possible that dietary intake of cruciferous vegetables including glucoraphanin (or SFN) may prevent or minimize relapse from remission, induced by stress and/or inflammation in depressed patients. In the review, the author would like to discuss the role of Keap1-Nrf2 system in mood disorders.
Collapse
Affiliation(s)
- Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| |
Collapse
|
28
|
Bustamante AC, Armstrong DL, Uddin M. Epigenetic profiles associated with major depression in the human brain. Psychiatry Res 2018; 260:439-442. [PMID: 29272728 DOI: 10.1016/j.psychres.2017.12.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/01/2017] [Accepted: 12/06/2017] [Indexed: 01/25/2023]
Abstract
We conducted an epigenome-wide association study of Major Depressive Disorder (MDD) in brain-derived DNA using two analytic approaches. DNA methylation data (GSE41826) was used in differential methylation (DM) analyses controlling for age, sex, suicide status, and post-mortem interval; and in weighted gene co-methylation network analyses (WGCNA) in probes mapping to transcription start sites. No probes in the DM analysis survived FDR correction. Nominally significant DM probes were enriched in synaptic function-related genes. WGCNA revealed one module correlated with MDD, enriched in genes associated with mitochondrial function. DM and WGCNA both showed enrichment of genes involved in transcription and DNA binding.
Collapse
Affiliation(s)
- Angela C Bustamante
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Don L Armstrong
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Monica Uddin
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA; Department of Psychology, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
| |
Collapse
|
29
|
Levine ME, Crimmins EM, Weir DR, Cole SW. Contemporaneous Social Environment and the Architecture of Late-Life Gene Expression Profiles. Am J Epidemiol 2017; 186:503-509. [PMID: 28911009 DOI: 10.1093/aje/kwx147] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 12/22/2016] [Indexed: 12/13/2022] Open
Abstract
Environmental or social challenges can stimulate a cascade of coordinated physiological changes in stress response systems. Unfortunately, chronic activation of these adaptations under conditions such as low socioeconomic status (SES) can have negative consequences for long-term health. While there is substantial evidence tying low SES to increased disease risk and reduced life expectancy, the underlying biology remains poorly understood. Using pilot data on 120 older adults from the Health and Retirement Study (United States, 2002-2010), we examined the associations between SES and gene expression levels in adulthood, with particular focus on a gene expression program known as the conserved transcriptional response to adversity. We also used a bioinformatics-based approach to assess the activity of specific gene regulation pathways involved in inflammation, antiviral responses, and stress-related neuroendocrine signaling. We found that low SES was related to increased expression of conserved transcriptional response to adversity genes and distinct patterns of proinflammatory, antiviral, and stress signaling (e.g., sympathetic nervous system and hypothalamic-pituitary-adrenal axis) transcription factor activation.
Collapse
Affiliation(s)
- Morgan E Levine
- Department of Human Genetics, Gonda Research Center, David Geffen School of Medicine, University of California, Los Angeles, USA
| | | | | | | |
Collapse
|
30
|
Steenkamp LR, Hough CM, Reus VI, Jain FA, Epel ES, James SJ, Morford AE, Mellon SH, Wolkowitz OM, Lindqvist D. Severity of anxiety- but not depression- is associated with oxidative stress in Major Depressive Disorder. J Affect Disord 2017; 219:193-200. [PMID: 28564628 PMCID: PMC5550320 DOI: 10.1016/j.jad.2017.04.042] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/22/2017] [Accepted: 04/23/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Oxidative stress is implicated in both depression and anxiety, but it is currently unclear whether this relates to syndromal diagnoses or trans-diagnostic dimensional symptoms. We examined the relationship between oxidative stress and severity of depression and anxiety symptoms in individuals with Major Depressive Disorder (MDD). METHODS Plasma oxidative stress markers F2-isoprostanes and oxidized glutathione (GSSG), and the antioxidant reduced glutathione (GSH), were assessed in 69 physically healthy, medication-free MDD subjects. Symptoms of anxiety and depression were assessed using the Hamilton Anxiety (HAM-A) and Hamilton Depression (HAM-D) Rating Scales. Total HAM-A and HAM-D scores, along with "core" anxiety and depression subscales, and individual HAM-D items "psychic anxiety" and "depressed mood," were related to oxidative stress markers. Analyses controlled for age, sex, BMI, and smoking. RESULTS Total HAM-A ratings were positively associated with F2-isoprostanes (β=.26, p=.042) and GSSG (β=.25, p=.049), but not GSH (β=.05, p=.711). Core anxiety severity was positively associated with F2-isoprostanes (β=.34, p=.012) and GSSG, although this did not reach significance (β=.24, p=.074). None of the biological markers were significantly associated with total HAM-D or core depression ratings (all p>.13). Subjects scoring high on "psychic anxiety" had elevated F2-isoprostanes (p=.030) and GSSG (p=.020). This was not seen with "depressed mood" scores (all p>.12). LIMITATIONS We assessed peripheral oxidative markers, but their relationship to the brain is unclear. CONCLUSIONS Oxidative stress is more closely related to anxiety than depression symptoms in MDD. This highlights the importance of relating oxidative stress to specific symptoms and could provide new insights into the biological correlates of affective disorders.
Collapse
Affiliation(s)
- Lisa R. Steenkamp
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America,Institute of Psychology, Leiden University, Leiden, the Netherlands
| | - Christina M. Hough
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - Victor I. Reus
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - Felipe A. Jain
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - Elissa S. Epel
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - S. Jill James
- Arkansas Children's Research Institute, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Alexandra E. Morford
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - Synthia H. Mellon
- Department of OB/GYN and Reproductive Sciences, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - Owen M. Wolkowitz
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America,Corresponding author: Owen M. Wolkowitz, MD, PhD, Department of Psychiatry, University of California, San Francisco (UCSF), School of Medicine, 401 Parnassus Avenue, San Francisco, CA 94143, USA, Phone: 415-476-7433,
| | - Daniel Lindqvist
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America,Lund University, Faculty of Medicine, Department of Clinical Sciences, Lund, Psychiatry, Sweden
| |
Collapse
|
31
|
Lindqvist D, Dhabhar FS, James SJ, Hough CM, Jain FA, Bersani FS, Reus VI, Verhoeven JE, Epel ES, Mahan L, Rosser R, Wolkowitz OM, Mellon SH. Oxidative stress, inflammation and treatment response in major depression. Psychoneuroendocrinology 2017; 76:197-205. [PMID: 27960139 PMCID: PMC5272818 DOI: 10.1016/j.psyneuen.2016.11.031] [Citation(s) in RCA: 327] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/24/2016] [Accepted: 11/28/2016] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Increased inflammation and oxidative stress have been shown in Major Depressive Disorder (MDD), although there is significant heterogeneity across studies. Whether markers of inflammation and oxidative stress are associated with antidepressant treatment response in MDD is currently unclear. The goals of the present study are to investigate markers of inflammation and oxidative stress in unmedicated MDD subjects and controls and test the relationship between these markers and antidepressant response in MDD subjects. METHODS Interleukin (IL)-6, tumor necrosis factor (TNF)-α, C-reactive protein, F2-isoprostanes, 8-OH 2-deoxyguanosine (8-OHdG), glutathione peroxidase, glutathione, and vitamin C were quantified in blood samples from 50 unmedicated MDD subjects and 55 healthy controls. Depression symptom severity was rated with the 17-item Hamilton Depression Rating Scale (HDRS). All subjects were somatically healthy and free from medications that could interfere with inflammation and oxidative stress markers. A subgroup of 22 MDD subjects underwent open-label selective serotonin reuptake inhibitor (SSRI) antidepressant treatment for eight weeks, after which blood sampling and the HDRS were repeated. Antidepressant treatment "response" was defined as ≥50% decrease in HDRS ratings over 8 weeks of treatment. RESULTS After controlling for the effects of age, sex, body mass index and smoking, MDD subjects had significantly higher levels of IL-6 (p<0.001), TNF-α (p<0.001), 8-OHdG (p=0.018), and F2-isoprostanes (p=0.012). Compared to Responders, Non-responders to SSRI antidepressant treatment had higher levels of F2-isoprostanes at baseline (p=0.006), and after eight weeks of treatment (p=0.031). Non-responders showed a significant increase in 8-OHdG over the course of treatment (p=0.021), whereas Responders showed a significant decrease in IL-6 over the course of treatment (p=0.019). CONCLUSION Our results are in line with previous reports of increased levels of markers of inflammation and oxidative stress in MDD. Moreover, poorer antidepressant treatment response was related to higher baseline levels of the major oxidative stress marker, F2-isoprostanes, in vivo. Further, antidepressant response was associated with changes in oxidative (8-OHdG) and inflammatory (IL-6) markers.
Collapse
Affiliation(s)
- Daniel Lindqvist
- Department of Psychiatry, UCSF Weill Institute for Neurosciences, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, United States; Lund University, Faculty of Medicine, Department of Clinical Sciences, Psychiatry, Lund, Sweden.
| | - Firdaus S. Dhabhar
- Department of Psychiatry & Behavioral Sciences, Sylvester Comprehensive Cancer Center, University of Miami, Florida, United States of America
| | - S. Jill James
- Arkansas Children's Research Institute, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Christina M. Hough
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - Felipe A. Jain
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - F. Saverio Bersani
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America,Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy
| | - Victor I. Reus
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - Josine E. Verhoeven
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America,Department of Psychiatry and EMGO Institute for Health and Care Research, VU University Medical Centre, Amsterdam, The Netherlands
| | - Elissa S. Epel
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - Laura Mahan
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - Rebecca Rosser
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - Owen M. Wolkowitz
- Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| | - Synthia H. Mellon
- Department of OB/GYN and Reproductive Sciences, University of California San Francisco (UCSF) School of Medicine, San Francisco, California, United States of America
| |
Collapse
|
32
|
Kovanen L, Donner K, Kaunisto M, Partonen T. PRKCDBP (CAVIN3) and CRY2 associate with major depressive disorder. J Affect Disord 2017; 207:136-140. [PMID: 27721187 DOI: 10.1016/j.jad.2016.09.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/12/2016] [Accepted: 09/25/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND Dysfunctions in the intrinsic clocks are suggested in patients with depressive disorders. The cryptochrome circadian clocks 1 and 2 (CRY1 and CRY2) proteins modulate circadian rhythms in a cell and influence emotional reactions and mood in an individual. The protein kinase C delta binding protein (PRKCDBP, or CAVIN3), similar to the serum deprivation response protein (SDPR, or CAVIN2), reduces metabolic stability of the PER2-CRY2 transcription factor complex that plays a role in the circadian rhythm synchronization. Our aim was to study SDPR, PRKCDBP, CRY1 and CRY2 genetic variants in depressive disorders. METHODS The sample included 5910 Finnish individuals assessed with the Munich-Composite International Diagnostic Interview (M-CIDI) in year 2000. In year 2011, 3424 individuals were assessed again. After genotype quality control, there were 383 subjects with major depressive disorder, 166 with dysthymia, and 479 with depressive disorders (major depressive disorder, dysthymia or both), and 4154 healthy controls. A total of 48 single-nucleotide polymorphisms from SDPR, PRKCDBP, CRY1 and CRY2 genes were analyzed using logistic regression models controlling for age and gender. RESULTS The earlier reported association of CRY2 variants with dysthymia was confirmed and extended to major depressive disorder (q<0.05). In addition, novel associations of PRKCDBP rs1488864 with depressive disorders (q=0.02) and with major depressive disorder in specific (q=0.007) were found. LIMITATIONS The number of cases was moderate and coverage of PRKCDB was limited. CONCLUSIONS CRY2 and PRKCDBP variants may be risk factors of major depressive disorder and provide information for diagnosis.
Collapse
Affiliation(s)
- Leena Kovanen
- Department of Health, National Institute for Health and Welfare (THL), Helsinki, Finland.
| | - Kati Donner
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Mari Kaunisto
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Timo Partonen
- Department of Health, National Institute for Health and Welfare (THL), Helsinki, Finland
| |
Collapse
|