1
|
Venable KE, Lee CC, Francis J. Addressing Mental Health in Rural Settings: A Narrative Review of Blueberry Supplementation as a Natural Intervention. Nutrients 2024; 16:3539. [PMID: 39458533 PMCID: PMC11510281 DOI: 10.3390/nu16203539] [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: 08/14/2024] [Revised: 09/23/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Depression and anxiety are major public health issues; however, there is an unmet need for novel, effective, and accessible treatments, particularly in rural communities. Blueberries are an unexplored nutraceutical for these conditions due to their excellent nutritional profile, with particularly high levels of polyphenols and anthocyanins and benefits on mood, cognition, and health. Here, we present a narrative review of the literature concerning the etiology and treatments of major depressive disorder (MDD) and generalized anxiety disorder (GAD). In both animal and human studies, blueberry supplementation can ameliorate behavioral symptoms of both anxiety and depression. The mechanistic underpinnings of these behavioral improvements are not fully defined, but likely involve biochemical alterations in the gut-brain axis, including to inflammatory cytokines, reactive oxygen species, and growth factors. We also review the limitations of traditional therapies in rural settings. Finally, we assess the potential benefit of nutraceutical interventions, particularly blueberries, as novel therapeutics for these distinct, yet related mental health issues.
Collapse
Affiliation(s)
- Katy E. Venable
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA; (C.C.L.); (J.F.)
| | | | | |
Collapse
|
2
|
Meyer JH, Wang Z, Santhirakumar A, Dowlati Y, Docteur N, Shoaib A, Purnava J, Wang Y, Wang W, Chen S, Husain MI, de Silva Wijeyeratne R, Reeyaz H, Baena-Tan C, Koshimori Y, Nasser Z, Sit V. Dietary supplement for mood symptoms in early postpartum: a double-blind randomized placebo controlled trial. EClinicalMedicine 2024; 71:102593. [PMID: 38813444 PMCID: PMC11133796 DOI: 10.1016/j.eclinm.2024.102593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 05/31/2024] Open
Abstract
Background Postpartum blues (PPB) is a frequent syndrome of sad mood, crying spells, anxiety, restlessness, reduced appetite, and irritability, typically peaking day 5 postpartum. When severe, it greatly increases risk for later postpartum depression. This trial compared a dietary supplement to placebo on PPB severity. The supplement was designed to counter downstream effects of elevated monoamine oxidase A level, implicated in causing PPB. Methods Participants recruited by advertisement from the Toronto region completed procedures at CAMH, Canada and/or participants' homes. Oral supplement or identical appearing relatively inert placebo were administered in randomised, double-blind fashion. Supplement was blueberry juice and extract given four times between nighttime day 3 and morning day 5 postpartum; tryptophan 2 g nighttime day 4 postpartum, and tyrosine 10 g morning day 5 postpartum. On day 5, depressed mood induction procedure (MIP) and postpartum blues were assessed. All data is presented (NCT03296956 closed, clinicaltrials.gov). Findings Between January 2019 and December 2022, participants took supplement (n = 51) or placebo (n = 52). There was no significant effect on primary outcome MIP on visual analogue scale for depressed mood (mean difference = -0.39 mm, 95% CI: -6.42 to 5.65 mm). Stein Maternity Blues scores, exploratory PPB measure, was lower in the active group (effect size 0.62; median, interquartile range (IQR): active 2.00 (IQR 1, 4); placebo 4.00 (IQR 1.5, 6); regression with general linear model, supplement effect, β coefficient = -1.50 (95%: CI -2.60, -0.40), p = 0.008; effect of CES-D crying category before supplement, p = 0.03-0.00000023). Twenty-six and 40 different adverse events occurred within 25% and 42% of supplement and placebo cases respectively (Chi-Square, p = 0.06). Interpretation The primary outcome was negative for effect on depressed mood induction, however the supplement moderately reduced PPB. Funding CAMH/Exeltis.
Collapse
Affiliation(s)
- Jeffrey H. Meyer
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - ZhaoHui Wang
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Apitharani Santhirakumar
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Yekta Dowlati
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Natalia Docteur
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Aqsa Shoaib
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Jareeat Purnava
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Yanqi Wang
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Wei Wang
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Sheng Chen
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Muhammad I. Husain
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Rashmi de Silva Wijeyeratne
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Heba Reeyaz
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Catalina Baena-Tan
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Yuko Koshimori
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Zahra Nasser
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| | - Valery Sit
- Brain Health Imaging Centre, Azrieli Centre for Neuro-Radiochemistry, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), 250 College Street, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College Street, 8th Floor, Toronto, M5T 1R8, Canada
| |
Collapse
|
3
|
Colombage RL, Holden S, Lamport DJ, Barfoot KL. The effects of flavonoid supplementation on the mental health of postpartum parents. Front Glob Womens Health 2024; 5:1345353. [PMID: 38577523 PMCID: PMC10993701 DOI: 10.3389/fgwh.2024.1345353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/27/2024] [Indexed: 04/06/2024] Open
Abstract
Introduction During the postpartum period, parents face psychological challenges and consequently, changes in mood and associated mood disorders have become increasingly prevalent in the 6-months following birth. Dietary flavonoids have been found to benefit mood and are therefore an appealing non-pharmacological option for potentially treating mood disorders in the postpartum. The aim of this study was to investigate whether a two-week dietary flavonoid intervention would improve mothers' and fathers' mental health in the immediate 6-month postpartum period. Method The study employed a randomised, parallel groups, controlled design to explore the effects of a flavonoid intervention vs. control group on several outcomes, including mood (PANAS), postpartum depression (EPDS), postpartum anxiety (PSAS-RSF-C) and quality of life (WHOQOL). Sixty participants (mothers n = 40, fathers n = 20) in the 6-month post-partum period were randomised to either a "flavonoid" or "control" condition. The flavonoid group were asked to add two flavonoid-rich foods (approximate flavonoid intake 218 mg/day) into their daily diet whilst controls (n = 23) were asked to continue with their usual diet for two-weeks (ClinicalTrials.gov (NCT04990622). Results Significant effects were found in the flavonoid group where mothers reported higher positive affect and lower postpartum depression after the two-week intervention relative to baseline. This finding is especially relevant as a clinical reduction in postpartum depression scores in the flavonoid group by an average 2.6 scoring points was observed, which equated to a reduction from "possible depression" at baseline to "little or no depression" at 2-weeks, which was not observed in the control group. Fathers' data was not analysed due to non-compliance with the intervention. Discussion This study provides evidence for the benefits of a dietary flavonoid intervention for mood and mental health in new mothers, supporting the utility of non-pharmacological, self-administrable changes to the diet for improving positive mood outcomes and reducing symptoms of postpartum depression in mothers during an especially challenging time. Further research for the effect of dietary interventions on paternal mental health is needed. Clinical Trial Registration ClinicalTrials.gov, identifier NCT04990622.
Collapse
Affiliation(s)
| | | | | | - Katie Louise Barfoot
- School of Psychology and Clinical Language Sciences, University of Reading, Reading, United Kingdom
| |
Collapse
|
4
|
Yan Z, Xu Y, Li K, Liu L. Increased fruit intake is associated with reduced risk of depression: evidence from cross-sectional and Mendelian randomization analyses. Front Public Health 2023; 11:1276326. [PMID: 38155897 PMCID: PMC10753833 DOI: 10.3389/fpubh.2023.1276326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/27/2023] [Indexed: 12/30/2023] Open
Abstract
Background The association between dietary patterns and depression has gained significant attention, but the relationship between fruit intake and depression remains unclear. This study aimed to investigate the role of fruit intake in the risk of depression using data from the National Health and Nutrition Examination Survey (NHANES) and Mendelian randomization (MR) analysis, and further explore the causal relationship between them. Materials and methods Cross-sectional analysis was conducted using the 2005-2018 NHANES data. Specialized weighted complex survey design analysis software was used for multivariate logistic analysis. Additionally, genetic variants for fruit intake and depression, as well as its related neuroticism traits, from the GWAS were used as instrumental variables in MR analysis. The inverse variance weighted (IVW) method was employed as the primary analysis method to evaluate the causal relationship between them. MR-Egger regression, MR-PRESSO test, and leave-one-out analysis were conducted to assess heterogeneity and pleiotropy. Results In NHANES, compared to the lowest quartile (Q1, <0.12 cup], the highest quartile (Q4, >1.49 cups) of fruit intake showed a significant reduction in the risk of depression after adjusting for relevant covariates. Model 3, after rigorous adjustment for multiple covariates, demonstrated improved predictive performance in both Receiver operating characteristic (ROC) curve and Decision curve analysis (DCA). In Model 3, the proportion of reduced depression risk associated with fruit intake reached 31% (OR = 0.69, 95% CI: 0.50-0.95). This association remained significant in the MR analysis (OR = 0.92, 95% CI = 0.87-0.96; p = 5.09E-04). Fruit intake was also associated with a decreased risk of neuroticism traits related to depression, including feeling lonely (OR = 0.82, 95% CI = 0.74-0.90; p = 2.91E-05), feeling miserable (OR = 0.79, 95% CI = 0.72-0.87; p = 2.35E-06), feeling fed-up (OR = 0.75, 95% CI = 0.68-0.83; p = 2.78E-08), irritable mood (OR = 0.89, 95% CI = 0.79-0.99; p = 0.03), and neuroticism (OR = 0.85, 95% CI = 0.76-0.96; p = 9.94E-03). The causal relationship between feeling lonely and fruit intake was bidirectional. Conclusion Increased fruit intake has a causal effect in reducing the risk of depression and is beneficial for related psychological well-being.
Collapse
Affiliation(s)
- Zhaoqi Yan
- Graduate School, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Yifeng Xu
- Graduate School, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Keke Li
- Graduate School, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| | - Liangji Liu
- Department of Respiratory and Critical Care Medicine, Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
5
|
Toh MPS, Yang CY, Lim PC, Loh HLJ, Bergonzelli G, Lavalle L, Mardhy E, Samuel TM, Suniega-Tolentino E, Silva Zolezzi I, Fries LR, Chan SY. A Probiotic Intervention With Bifidobacterium longum NCC3001 on Perinatal Mood Outcomes (PROMOTE Study): Protocol for a Decentralized Randomized Controlled Trial. JMIR Res Protoc 2023; 12:e41751. [PMID: 37018024 PMCID: PMC10131660 DOI: 10.2196/41751] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 02/07/2023] [Accepted: 02/07/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Perinatal mood disorders such as depression and anxiety are common, with subclinical symptomology manifesting as perinatal mood disturbances being even more prevalent. These could potentially affect breastfeeding practices and infant development. Pregnant and lactating women usually limit their exposure to medications, including those for psychological symptoms. Interestingly, the naturally occurring probiotic Bifidobacterium longum (BL) NCC3001 has been shown to reduce anxious behavior in preclinical models and feelings of low mood in nonpregnant human adults. During the COVID-19 pandemic, mental health issues increased, and conventionally conducted clinical trials were restricted by social distancing regulations. OBJECTIVE This study, Probiotics on Mothers' Mood and Stress (PROMOTE), aimed to use a decentralized clinical trial design to test whether BL NCC3001 can reduce symptoms of depression, anxiety, and stress over the perinatal period. METHODS This double-blind, placebo-controlled, randomized, and 3-parallel-arm study aimed to recruit 180 women to evaluate the efficacy of the probiotic taken either during pregnancy and post partum (from 28-32 weeks' gestation until 12 weeks after delivery; n=60, 33.3%) or post partum only (from birth until 12 weeks after delivery; n=60, 33.3%) in comparison with a placebo control group (n=60, 33.3%). Participants consumed the probiotic or matched placebo in a drink once daily. Mood outcomes were measured using the State-Trait Anxiety Inventory and Edinburgh Postnatal Depression Scale questionnaires, captured electronically at baseline (28-32 weeks' gestation) and during e-study sessions over 5 further time points (36 weeks' gestation; 9 days post partum; and 4, 8, and 12 weeks post partum). Saliva and stool samples were collected longitudinally at home to provide mechanistic insights. RESULTS In total, 520 women registered their interest on our website, of whom 184 (35.4%) were eligible and randomized. Of these 184 participants, 5 (2.7%) withdrew after randomization, leaving 179 (97.3%) who completed the study. Recruitment occurred between November 7, 2020, and August 20, 2021. Advertising on social media brought in 46.9% (244/520) of the prospective participants, followed by parenting-specific websites (116/520, 22.3%). Nationwide recruitment was achieved. Data processing is ongoing, and there are no outcomes to report yet. CONCLUSIONS Multiple converging factors contributed to speedy recruitment and retention of participants despite COVID-19-related restrictions. This decentralized trial design sets a precedent for similar studies, in addition to potentially providing novel evidence on the impact of BL NCC3001 on symptoms of perinatal mood disturbances. This study was ideal for remote conduct: because of the high digital literacy and public trust in digital security in Singapore, the intervention could be self-administered without regular clinical monitoring, and the eligibility criteria and outcomes were measured using electronic questionnaires and self-collected biological samples. This design was particularly suited for a group considered vulnerable-pregnant women-during the challenging times of COVID-19-related social restrictions. TRIAL REGISTRATION ClinicalTrials.gov NCT04685252; https://clinicaltrials.gov/ct2/show/NCT04685252. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/41751.
Collapse
Affiliation(s)
- Melissa P S Toh
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| | - Chui Yuen Yang
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| | - Phei Cze Lim
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| | - Hui Li J Loh
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| | | | - Luca Lavalle
- Nestle Research, Lausanne, Lausanne, Switzerland
| | - Elias Mardhy
- Nestle Research, Lausanne, Lausanne, Switzerland
| | | | | | | | | | - Shiao Yng Chan
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
6
|
Tsai Z, Shah N, Tahir U, Mortaji N, Owais S, Perreault M, Van Lieshout RJ. Dietary interventions for perinatal depression and anxiety: a systematic review and meta-analysis of randomized controlled trials. Am J Clin Nutr 2023:S0002-9165(23)46315-0. [PMID: 37019362 DOI: 10.1016/j.ajcnut.2023.03.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Dietary interventions are a widely available intervention for depression and anxiety among pregnant and/or postpartum (i.e., perinatal) persons but their effectiveness is not well known. OBJECTIVE We performed a systematic review and meta-analysis to assess the effectiveness of dietary interventions for the treatment of perinatal depression and/or anxiety. DESIGN We searched Medline, EMBASE, PsycINFO, CINAHL, and Web of Science from their inception to November 2, 2022. Studies were included if they were available in English and examined the effectiveness of a dietary intervention for perinatal depression and/or anxiety in a randomized controlled trial. RESULTS Our search identified 4,246 articles, 36 of which were included and 28 were eligible for meta-analysis. Random effects meta-analyses were performed. Polyunsaturated fatty acids (PUFAs) were not found to improve symptoms of perinatal depression compared to control conditions (SMD -0.11; 95% CI -0.26 to 0.04). These results did not change when examined during pregnancy or the postpartum period separately, nor did they vary according to fatty acid ratio. Elemental metals (iron, zinc, and magnesium) were also not found to be superior to placebo (SMD, -0.42; 95% CI, -1.05 to 0.21), though, vitamin D yielded a small to medium effect size improvements (SMD, -0.52; 95% CI, -0.84 to -0.20) in postpartum depression. Iron may help in those with confirmed iron deficiency. Narrative synthesis was performed for studies ineligible for meta-analyses. CONCLUSIONS Despite their widespread popularity, PUFAs and elemental metals do not appear to effectively reduce perinatal depression. Vitamin D taken in doses of 1800 to 3500 International Units (IU) per day may have some promise. Additional high-quality, large-scale randomized controlled trials (RCTs) are needed to determine the true effectiveness of dietary interventions on perinatal depression and/or anxiety. PROSPERO REGISTRATION DATE AND NUMBER July 5th 2020, CRD42020208830.
Collapse
Affiliation(s)
- Zoe Tsai
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - Nirmay Shah
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Umair Tahir
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Neda Mortaji
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Sawayra Owais
- Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Maude Perreault
- Faculty of Medicine, University of Montreal, Montreal, Québec, Canada
| | - Ryan J Van Lieshout
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
7
|
Høgh S, Hegaard HK, Renault KM, Cvetanovska E, Kjærbye-Thygesen A, Juul A, Borgsted C, Bjertrup AJ, Miskowiak KW, Væver MS, Stenbæk DS, Dam VH, Binder E, Ozenne B, Mehta D, Frokjaer VG. Short-term oestrogen as a strategy to prevent postpartum depression in high-risk women: protocol for the double-blind, randomised, placebo-controlled MAMA clinical trial. BMJ Open 2021; 11:e052922. [PMID: 35763351 PMCID: PMC8719185 DOI: 10.1136/bmjopen-2021-052922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Postpartum depression affects 10%-15% of women and has a recurrence rate of 40% in subsequent pregnancies. Women who develop postpartum depression are suspected to be more sensitive to the rapid and large fluctuations in sex steroid hormones, particularly estradiol, during pregnancy and postpartum. This trial aims to evaluate the preventive effect of 3 weeks transdermal estradiol treatment immediately postpartum on depressive episodes in women at high risk for developing postpartum depression. METHODS AND ANALYSIS The Maternal Mental Health Trial is a double-blind, randomised and placebo-controlled clinical trial. The trial involves three departments of obstetrics organised under Copenhagen University Hospital in Denmark. Women who are singleton pregnant with a history of perinatal depression are eligible to participate. Participants will be randomised to receive either transdermal estradiol patches (200 µg/day) or placebo patches for 3 weeks immediately postpartum. The primary outcome is clinical depression, according to the Diagnostic and Statistical Manual of Mental Disorders-V criteria of Major Depressive Disorder with onset at any time between 0 and 6 months postpartum. Secondary outcomes include, but are not limited to, symptoms of depression postpartum, exclusive breastfeeding, cortisol dynamics, maternal distress sensitivity and cognitive function. The primary statistical analysis will be performed based on the intention-to-treat principle. With the inclusion of 220 participants and a 20% expected dropout rate, we anticipate 80% power to detect a 50% reduction in postpartum depressive episodes while controlling the type 1 error at 5%. ETHICS AND DISSEMINATION The study protocol is approved by the Regional Committees on Health Research Ethics in the Capital Region of Denmark, the Danish Medicines Agency and the Centre for Data Protection Compliance in the Capital Region of Denmark. We will present results at scientific meetings and in peer-reviewed journals and in other formats to engage policymakers and the public. TRIAL REGISTRATION NUMBER NCT04685148.
Collapse
Affiliation(s)
- Stinne Høgh
- Department of Obstetrics and Gynaecology, Rigshospitalet, Copenhagen, Denmark
- Neurobiology Research Unit, Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | - Anders Juul
- Department of Growth and Reproduction, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Camilla Borgsted
- Neurobiology Research Unit, Rigshospitalet, Copenhagen, Denmark
- Department of Psychiatry, Rigshospitalet, Copenhagen, Denmark
| | | | - Kamilla Woznica Miskowiak
- Department of Psychiatry, Rigshospitalet, Copenhagen, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | | | - Dea Siggaard Stenbæk
- Neurobiology Research Unit, Rigshospitalet, Copenhagen, Denmark
- Department of Psychology, University of Copenhagen, Copenhagen, Denmark
| | | | - Elisabeth Binder
- Department of Translational Research in Psychiatry, Max-Planck-Institute for Psychiatry, Munchen, Bayern, Germany
| | - Brice Ozenne
- Neurobiology Research Unit, Rigshospitalet, Copenhagen, Denmark
| | - Divya Mehta
- Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Vibe G Frokjaer
- Neurobiology Research Unit, Rigshospitalet, Copenhagen, Denmark
- Department of Psychiatry, Rigshospitalet, Copenhagen, Denmark
- Capital Region of Denmark Mental Health Services, Copenhagen, Denmark
| |
Collapse
|
8
|
Mental Health in New Mothers: A Randomised Controlled Study into the Effects of Dietary Flavonoids on Mood and Perceived Quality of Life. Nutrients 2021; 13:nu13072383. [PMID: 34371893 PMCID: PMC8308795 DOI: 10.3390/nu13072383] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/10/2021] [Indexed: 12/27/2022] Open
Abstract
The postnatal period is a significant period of physical, physiological and psychological change for mothers, rendering them particularly vulnerable to changes in mood or disorders such as postnatal depression (PND). Previous interventions with foods high in flavonoids have demonstrated beneficial acute and chronic mood effects in healthy child, adolescent and adult populations. It is unclear whether mood effects persist in populations who are potentially at-risk of developing mood disorders, such as postnatal mothers. This exploratory study investigated the effects of a 2-week daily dietary flavonoid intervention on mood (PANAS-NOW), anxiety (STAI), depressive symptoms (PHQ-8) and perceived quality of life (WHOQOL-BREF) in forty-one new mothers in the 0–12-month postnatal period, before and after flavonoid intervention. Mothers either added high flavonoid foods to their daily diet, or did not include additions following a randomised, between-groups, controlled design. Significant effects were observed in the flavonoid group with mothers reporting lower state anxiety and higher perceived quality of physical health at the 2-week timepoint. These findings suggest that regular dietary consumption of flavonoids may benefit mothers’ anxiety and perceived quality of life in the postnatal period. Replication of these results may indicate the potential for dietary flavonoids to promote healthy mood regulation in mothers or prevent the onset or severity of symptoms in postnatal psychological disorders, both of which would be beneficial for women’s health services and public mental health.
Collapse
|
9
|
Wu Z, Zhou L, Sun L, Xie Y, Xiao L, Wang H, Wang G. Brief postpartum separation from offspring promotes resilience to lipopolysaccharide challenge-induced anxiety and depressive-like behaviors and inhibits neuroinflammation in C57BL/6J dams. Brain Behav Immun 2021; 95:190-202. [PMID: 33766700 DOI: 10.1016/j.bbi.2021.03.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 02/25/2021] [Accepted: 03/17/2021] [Indexed: 12/27/2022] Open
Abstract
Emerging evidence indicates an important role for neuroinflammation in depression. Brief maternal separation promotes resilience to depression in offspring, but relatively little is known about the effects of different durations of postpartum separation (PS) from offspring on anxiety and depressive-like behaviors in dams following immune challenge. Lactating C57BL/6J mice were subjected to no separation (NPS), brief PS (15 min/day, PS15) or prolonged PS (180 min/day, PS180) from postpartum day (PPD) 1 to PPD21 and then injected with lipopolysaccharide (LPS). Behavioral tests, including the open field test (OFT) and forced swimming test (FST), were carried out at 24 h after the injection. LPSresulted in anxiety and depressive-like behaviors in NPS dams and activated ionized calcium-binding adaptor molecule (Iba1), an important biomarker of microglia, in the hippocampus. However, compared with NPS + LPS dams, PS15 + LPS dams spent significantly more time in the center of the OFT (anxiety-like behavior) and exhibited lower immobility time in the FST (depressive-like behavior), which indicated a phenomenon of resilience. Furthermore, the activation of neuroinflammation was inhibited in PS15 dams. Specifically, levels of the Iba1 mRNA and protein were decreased, while the mRNA expression of NLR family pyrin domain containing 3 (NLRP3) inflammasome/interleukin-18 (IL-18)/nuclear factor kappa-B (NF-κB) was decreased in the hippocampus. Furthermore, positive linear correlations were observed between microglial activation and LPS-induced depressive-like behaviors in dams. Collectively, the findings of this study confirm that brief PS from offspring promotes resilience to LPS immune challenge-induced behavioral deficits and inhibits neuroinflammation in dams separated from their offspring during lactation.
Collapse
Affiliation(s)
- Zuotian Wu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan 430060, China
| | - Lin Zhou
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan 430060, China
| | - Limin Sun
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan 430060, China
| | - Yumeng Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan 430060, China
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan 430060, China
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan 430060, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan 430060, China.
| |
Collapse
|
10
|
Promising leads and pitfalls: a review of dietary supplements and hormone treatments to prevent postpartum blues and postpartum depression. Arch Womens Ment Health 2021; 24:381-389. [PMID: 33205315 DOI: 10.1007/s00737-020-01091-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/03/2020] [Indexed: 12/16/2022]
Abstract
Prevention of postpartum depression (PPD) is important because it typically has a 13% prevalence rate, impactful immediate symptoms with greater risk of suicide, and higher long-term risk of psychiatric symptoms in both the mother and family. There are no universal approaches across all childbearing women that have proven to be preventative for PPD, so it is hoped that dietary and/or hormonal interventions will be developed. There are some effective preventative approaches for PPD, such as psychotherapy and medical management, for the highest risk cases, like when there is a past history of a major depressive episode. The purpose is to review studies that assess dietary and hormonal interventions for prevention of PPD and/or postpartum blues, a high-risk state for PPD. Studies that assess dietary and hormonal interventions for prevention of PPD which included a comparison group were reviewed, including omega-3 fatty acids, mineral and vitamin supplements, amino acid combinations, allopregnanolone, progesterone, and thyroxine. Presently, development of dietary supplements and hormonal products for prevention of PPD is at an early stage with most trials showing results that are either preliminary, not definitive, trend level or variable across studies. Even so, a few directions are not recommended for further investigation such as progesterone and thyroxine. On the other hand, studies of allopregnanolone for prophylaxis of PPD are needed. Also, given the number of trend level findings and the multifactorial etiology of PPD, it may be prudent to investigate combined interventions rather than monotherapies. There is still a major need to develop a dietary supplement that creates resiliency against the biological changes in early postpartum associated with risk for mood disorders and/or PPD.
Collapse
|
11
|
Universal and Selective Interventions to Prevent Poor Mental Health Outcomes in Young People: Systematic Review and Meta-analysis. Harv Rev Psychiatry 2021; 29:196-215. [PMID: 33979106 DOI: 10.1097/hrp.0000000000000294] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Much is not known about the efficacy of interventions to prevent poor mental health outcomes in young people by targeting either the general population (universal prevention) or asymptomatic individuals with high risk of developing a mental disorder (selective prevention). METHODS We conducted a PRISMA/MOOSE-compliant systematic review and meta-analysis of Web of Science to identify studies comparing post-test efficacy (effect size [ES]; Hedges' g) of universal or selective interventions for poor mental health outcomes versus control groups, in samples with mean age <35 years (PROSPERO: CRD42018102143). Measurements included random-effects models, I2 statistics, publication bias, meta-regression, sensitivity analyses, quality assessments, number needed to treat, and population impact number. RESULTS 295 articles (447,206 individuals; mean age = 15.4) appraising 17 poor mental health outcomes were included. Compared to control conditions, universal and selective interventions improved (in descending magnitude order) interpersonal violence, general psychological distress, alcohol use, anxiety features, affective symptoms, other emotional and behavioral problems, consequences of alcohol use, posttraumatic stress disorder features, conduct problems, tobacco use, externalizing behaviors, attention-deficit/hyperactivity disorder features, and cannabis use, but not eating-related problems, impaired functioning, internalizing behavior, or sleep-related problems. Psychoeducation had the highest effect size for ADHD features, affective symptoms, and interpersonal violence. Psychotherapy had the highest effect size for anxiety features. CONCLUSION Universal and selective preventive interventions for young individuals are feasible and can improve poor mental health outcomes.
Collapse
|
12
|
Jones DN, Raghanti MA. The role of monoamine oxidase enzymes in the pathophysiology of neurological disorders. J Chem Neuroanat 2021; 114:101957. [PMID: 33836221 DOI: 10.1016/j.jchemneu.2021.101957] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/03/2021] [Accepted: 04/04/2021] [Indexed: 12/12/2022]
Abstract
Monoamine oxidase enzymes are responsible for the degredation of serotonin, dopamine, and norepinephrine in the central neurvous system. Although it has been nearly 100 years since they were first described, we are still learning about their role in the healthy brain and how they are altered in various disease states. The present review provides a survey of our current understanding of monoamine oxidases, with a focus on their contributions to neuropsychiatric, neurodevelopmental, and neurodegenerative disease. Important species differences in monoamine oxidase function and development in the brain are highlighted. Sex-specific monoamine oxidase regulatory mechanisms and their implications for various neurological disorders are also discussed. While our understanding of these critical enzymes has expanded over the last century, gaps exist in our understanding of sex and species differences and the roles monoamine oxidases may play in conditions often comorbid with neurological disorders.
Collapse
Affiliation(s)
- Danielle N Jones
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH, USA; Brain Health Research Institute, Kent State University, Kent, OH, USA.
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH, USA; Brain Health Research Institute, Kent State University, Kent, OH, USA
| |
Collapse
|
13
|
Sacher J, Chechko N, Dannlowski U, Walter M, Derntl B. The peripartum human brain: Current understanding and future perspectives. Front Neuroendocrinol 2020; 59:100859. [PMID: 32771399 DOI: 10.1016/j.yfrne.2020.100859] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022]
Abstract
The peripartum period offers a unique opportunity to improve our understanding of how dramatic fluctuations in endogenous ovarian hormones affect the human brain and behavior. This notwithstanding, peripartum depression remains an underdiagnosed and undertreated disorder. Here, we review recent neuroimaging findings with respect to the neuroplastic changes in the maternal brain during pregnancy and the postpartum period. We seek to provide an overview of multimodal neuroimaging designs of current peripartum depression models of hormone withdrawal, changes in monoaminergic signaling, and maladaptive neuroplasticity, which likely lead to the development of a condition that puts the lives of mother and infant at risk. We discuss the need to effectively integrate the available information on psychosocial and neurobiological risk factors contributing to individual vulnerability. Finally, we propose a systematic approach to neuroimaging the peripartum brain that acknowledges important co-morbidities and variation in disease onset.
Collapse
Affiliation(s)
- Julia Sacher
- Department of Neurology, Max-Planck-Institute for Human Cognitive and Brain Sciences, Stephanstr. 1A, 04103 Leipzig, Germany; Emotion Neuroimaging Lab, Max Planck Institute for Human Cognitive and Brain Sciences, Stephanstr. 1A, 04103 Leipzig, Germany; Clinic of Cognitive Neurology, University of Leipzig, Liebigstr. 16, 04103 Leipzig, Germany.
| | - Natalia Chechko
- Klinik für Psychiatrie, Psychotherapie und Psychosomatik, Pauwelsstr. 30, 52074 Aachen, Germany; Institute of Neuroscience and Medicine (INM-7: Brain and Behaviour), Forschungszentrum Jülich, Wilhelm-Johnen-Str., 52428 Jülich, Germany
| | - Udo Dannlowski
- Department of Psychiatry and Psychotherapy, University of Muenster, Albert Schweitzer-Campus 1, G 9A, 48149 Muenster, Germany
| | - Martin Walter
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Philosophenweg 3, 07743 Jena, Germany
| | - Birgit Derntl
- Department of Psychiatry and Psychotherapy, Medical School, University of Tübingen, Osianderstr. 24, 72076 Tübingen, Germany; LEAD Graduate Training & Research Network, University of Tübingen, Walter-Simon-Str. 12, 72072 Tübingen, Germany
| |
Collapse
|
14
|
Qiu W, Hodges TE, Clark EL, Blankers SA, Galea LAM. Perinatal depression: Heterogeneity of disease and in animal models. Front Neuroendocrinol 2020; 59:100854. [PMID: 32750403 DOI: 10.1016/j.yfrne.2020.100854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/14/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022]
Abstract
Perinatal depression (PND) can have either an antepartum or postpartum onset. Although the greatest risk factor for PND is previous depression history,de novoPND occurs with the majority of cases occurring in the postpartum. Timing of depression can impact etiology, prognosis, and response to treatment. Thus, it is crucial to study the impact of the heterogeneity of PND for better health outcomes. In this review, we outline the differences between antepartum and postpartum depression onset of PND. We discuss maternal physiological changes that differ between pregnancy and postpartum and how these may differentially impact depression susceptibility. We highlight changes in the maternal steroid and peptide hormone levels, immune signalling, serotonergic tone, metabolic factors, brain morphology, and the gut microbiome. Finally, we argue that studying the heterogeneity of PND in clinical and preclinical models can lead to improved knowledge of disease etiopathology and treatment outcomes.
Collapse
Affiliation(s)
- Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Travis E Hodges
- Department of Psychology, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Emily L Clark
- Graduate Program in Neuroscience, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Canada; Department of Psychology, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada.
| |
Collapse
|
15
|
Frokjaer VG. Pharmacological sex hormone manipulation as a risk model for depression. J Neurosci Res 2020; 98:1283-1292. [PMID: 32399989 PMCID: PMC7383584 DOI: 10.1002/jnr.24632] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 04/06/2020] [Accepted: 04/11/2020] [Indexed: 12/19/2022]
Abstract
Sex hormone transition may trigger severe depressive episodes in some women. In order to map mechanisms related to such phenomena we developed a pharmacological preclinical human model using sex hormone manipulation with gonadotropin releasing hormone agonist (GnRHa) in a placebo‐controlled design. Here the findings from this model is synthesized and discussed in the context of related literature on hormonal contributions to reproductive mental health disorders. The GnRha model work points to an estradiol‐dependent depressive response in healthy women undergoing short‐term sex hormone manipulation with GnRHa, which is linked to serotonin transporter changes (a key regulator of synaptic serotonin), a disengagement of hippocampus, and overengagement of brain networks recruited when processing emotional salient information. Further, the GnRHa model suggest that key brain regions in the reward circuit are less engaged in positive stimuli when undergoing sex hormone manipulation, which may underlie anhedonia. Also, the work supports that enhanced sensitivity to estrogen signaling at the level of gene expression may drive increased risk for depressive symptoms when exposed to sex steroid hormone fluctuations. In conclusion, the GnRHa model work highlights the brain signatures of rapid and profound changes in sex steroid hormone milieu, which reflect plausible mechanisms by which risk for mood disorders works. This model points to the role of estrogen dynamics and sensitivity, and offers a rationale for personalized prevention in hormonal transition phases, for example pregnancy to postpartum transition, perimenopause, and hormone treatments, which now can move into clinical translation and ideally pave the way for protecting mental and cognitive health.
Collapse
Affiliation(s)
- Vibe G Frokjaer
- Neurobiology Research Unit and Center for Integrated Molecular Brain Imaging, Copenhagen, Denmark.,Psychiatric Center Copenhagen, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
16
|
Stress, sex hormones, inflammation, and major depressive disorder: Extending Social Signal Transduction Theory of Depression to account for sex differences in mood disorders. Psychopharmacology (Berl) 2019; 236:3063-3079. [PMID: 31359117 PMCID: PMC6821593 DOI: 10.1007/s00213-019-05326-9] [Citation(s) in RCA: 226] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 07/08/2019] [Indexed: 12/11/2022]
Abstract
Social Signal Transduction Theory of Depression is a biologically plausible, multi-level theory that describes neural, physiologic, molecular, and genomic mechanisms that link experiences of social-environmental adversity with internal biological processes that drive depression pathogenesis, maintenance, and recurrence. Central to this theory is the hypothesis that interpersonal stressors involving social threat (e.g., social conflict, evaluation, rejection, isolation, and exclusion) upregulate inflammatory processes that can induce several depressive symptoms, including sad mood, anhedonia, fatigue, psychomotor retardation, and social-behavioral withdrawal. The original article describing this formulation (Psychol Bull 140:774-815, 2014) addressed critical questions involving depression onset and recurrence, as well as why depression is strongly predicted by early life stress and comorbid with anxiety disorders and certain physical disease conditions, such as asthma, rheumatoid arthritis, chronic pain, and cardiovascular disease. Here, we extend the theory to help explain sex differences in depression prevalence, which is a defining feature of this disorder. Central to this extension is research demonstrating that ovarian hormone fluctuations modulate women's susceptibility to stress, brain structure and function, and inflammatory activity and reactivity. These effects are evident at multiple levels and are highly context-dependent, varying as a function of several factors including sex, age, reproductive state, endogenous versus exogenous hormones, and hormone administration mode and dose. Together, these effects help explain why women are at greater risk for developing inflammation-related depressed mood and other neuropsychiatric, neurodevelopmental, and neurodegenerative disorders during the reproductive years, especially for those already at heightened risk for depression or in the midst of a hormonal transition period.
Collapse
|
17
|
Perinatal Depression: Embracing Variability toward Better Treatment and Outcomes. Neuron 2019; 102:13-16. [DOI: 10.1016/j.neuron.2019.02.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 12/13/2022]
|
18
|
Dowlati Y, de Jesus DR, Selby P, Fan I, Meyer JH. Depressed mood induction in early cigarette withdrawal is unaffected by acute monoamine precursor supplementation. Neuropsychiatr Dis Treat 2019; 15:311-321. [PMID: 30774343 PMCID: PMC6352866 DOI: 10.2147/ndt.s172334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Cigarette smoking is the leading preventable cause of death; however, quitting is difficult and early relapse is common. Dysphoric mood during early cigarette withdrawal is associated with relapse, and with the exception of bupropion and nortriptyline, few interventions have been developed to prevent this. During early cigarette withdrawal there is an elevation in the levels of monoamine oxidase-A (MAO-A), which removes monoamines excessively and induces oxidative stress and is implicated in creating sad mood. Hence, we conducted a randomized, placebo-controlled, double-blind crossover trial of a dietary supplement designed to counter the effects of elevated MAO-A levels on vulnerability to depressed mood. METHODS Twenty-one otherwise healthy cigarette smokers completed the protocol, receiving either active dietary supplement followed by washout and placebo or the same in reverse order. The dietary supplement was composed of monoamine precursors (2 g tryptophan, 10 g tyrosine) and blueberry antioxidants (blueberry juice with blueberry extract). Vulnerability to depressed mood was measured by the change in scores of depressed mood on the visual analog scale (VAS) following the sad mood induction paradigm (MIP). RESULTS There was a significant increase in VAS depressed mood scores after the sad MIP during supplement and placebo, but no difference between active and placebo conditions. There was also a significant increase in urge-to-smoke scores after sad MIP during supplement and placebo but no difference between active and placebo conditions. Reliability of the increase in VAS after MIP was very good. CONCLUSION The dietary supplement had negligible effect on depressed mood, but sad MIP is a very reliable method that can be applied in future studies to assess other interventions for preventing dysphoric mood during early cigarette withdrawal.
Collapse
Affiliation(s)
- Yekta Dowlati
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada, .,Department of Psychiatry, University of Toronto, ON, Canada,
| | - Danilo R de Jesus
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada, .,Department of Psychiatry, University of Toronto, ON, Canada,
| | - Peter Selby
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada, .,Department of Psychiatry, University of Toronto, ON, Canada, .,Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada
| | - Ian Fan
- Department of Psychiatry, University of Toronto, ON, Canada, .,Mood Disorders Association of British Columbia, Vancouver, BC, Canada
| | - Jeffrey H Meyer
- CAMH Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada, .,Department of Psychiatry, University of Toronto, ON, Canada,
| |
Collapse
|
19
|
Young SM, Gryder LK, Cross C, Zava D, Kimball DW, Benyshek DC. Placentophagy’s effects on mood, bonding, and fatigue: A pilot trial, part 2. Women Birth 2018; 31:e258-e271. [DOI: 10.1016/j.wombi.2017.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/04/2017] [Accepted: 11/04/2017] [Indexed: 10/18/2022]
|
20
|
Bharti R, Dey G, Das AK, Mandal M. Differential expression of IL-6/IL-6R and MAO-A regulates invasion/angiogenesis in breast cancer. Br J Cancer 2018; 118:1442-1452. [PMID: 29695771 PMCID: PMC5988749 DOI: 10.1038/s41416-018-0078-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 02/27/2018] [Accepted: 03/14/2018] [Indexed: 01/16/2023] Open
Abstract
Background Monoamine oxidases (MAO) are mitochondrial enzymes functioning in oxidative metabolism of monoamines. The action of MAO-A has been typically described in neuro-pharmacological domains. Here, we have established a co-relation between IL-6/IL-6R and MAO-A and their regulation in hypoxia induced invasion/angiogenesis. Methods We employed various in-vitro and in-vivo techniques and clinical samples. Results We studied a co-relation among MAO-A and IL-6/IL-6R and tumour angiogenesis/invasion in hypoxic environment in breast cancer model. Activation of IL-6/IL-6R and its downstream was found in hypoxic cancer cells. This elevation of IL-6/IL-6R caused sustained inhibition of MAO-A in hypoxic environment. Inhibition of IL-6R signalling or IL-6R siRNA increased MAO-A activity and inhibited tumour angiogenesis and invasion significantly in different models. Further, elevation of MAO-A with 5-azacytidine (5-Aza) modulated IL-6 mediated angiogenesis and invasive signatures including VEGF, MMPs and EMT in hypoxic breast cancer. High grade invasive ductal carcinoma (IDC) clinical specimen displayed elevated level of IL-6R and depleted MAO-A expression. Expression of VEGF and HIF-1α was unregulated and loss of E-Cadherin was observed in high grade IDC tissue specimen. Conclusions Suppression of MAO-A by IL-6/IL-6R activation promotes tumour angiogenesis and invasion in hypoxic breast cancer environment.
Collapse
Affiliation(s)
- Rashmi Bharti
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Goutam Dey
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Anjan Kumar Das
- Department of Pathology, Calcutta National Medical Collage, Kolkata, West Bengal, 70014, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, 721302, India.
| |
Collapse
|
21
|
Zsido RG, Villringer A, Sacher J. Using positron emission tomography to investigate hormone-mediated neurochemical changes across the female lifespan: implications for depression. Int Rev Psychiatry 2017; 29:580-596. [PMID: 29199875 DOI: 10.1080/09540261.2017.1397607] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Ovarian hormones, particularly oestrogen and progesterone, undergo major fluctuations across the female lifespan. These hormone transition periods, such as the transition from pregnancy to postpartum, as well as the transition into menopause (perimenopause), are also known to be times of elevated susceptibility to depression. This study reviews how these transition periods likely influence neurochemical changes in the brain that result in disease vulnerability. While there are known associations between oestrogen/progesterone and different monoaminergic systems, the interactions and their potential implications for mood disorders are relatively unknown. Positron Emission Tomography (PET) allows for the in-vivo quantification of such neurochemical changes, and, thus, can provide valuable insight into how both subtle and dramatic shifts in hormones contribute to the elevated rates of depression during pre-menstrual, post-partum, and perimenopausal periods in a woman's life. As one better understands how to address the challenges of PET studies involving highly vulnerable populations, such as women who have recently given birth, one will gain the insight necessary to design and individualize treatment and therapy. Understanding the precise time-line in younger women when dramatic fluctuations in the hormonal milieu may contribute to brain changes may present a powerful opportunity to intervene before a vulnerable state develops into a diseased state in later life.
Collapse
Affiliation(s)
- Rachel G Zsido
- a Emotion NeuroimaGinG(EGG)-Lab , Max Planck Institute for Cognitive and Brain Sciences , Leipzig , Germany.,b Department of Neurology , Max Planck Institute for Cognitive and Brain Sciences , Leipzig , Germany
| | - Arno Villringer
- b Department of Neurology , Max Planck Institute for Cognitive and Brain Sciences , Leipzig , Germany.,c Clinic for Cognitive Neurology , University of Leipzig , Leipzig , Germany
| | - Julia Sacher
- a Emotion NeuroimaGinG(EGG)-Lab , Max Planck Institute for Cognitive and Brain Sciences , Leipzig , Germany.,b Department of Neurology , Max Planck Institute for Cognitive and Brain Sciences , Leipzig , Germany.,c Clinic for Cognitive Neurology , University of Leipzig , Leipzig , Germany
| |
Collapse
|
22
|
Abstract
BACKGROUND Postpartum depression (PPD) could affect ~10% of women and impair the quality of mother-infant interactions. Currently, there are no objective methods to diagnose PPD. Therefore, this study was conducted to identify potential biomarkers for diagnosing PPD. MATERIALS AND METHODS Morning urine samples of PPD subjects, postpartum women without depression (PPWD) and healthy controls (HCs) were collected. The gas chromatography-mass spectroscopy (GC-MS)-based urinary metabolomic approach was performed to characterize the urinary metabolic profiling. The orthogonal partial least-squares-discriminant analysis (OPLS-DA) was used to identify the differential metabolites. The logistic regression analysis and Bayesian information criterion rule were further used to identify the potential biomarker panel. The receiver operating characteristic curve analysis was conducted to evaluate the diagnostic performance of the identified potential biomarker panel. RESULTS Totally, 73 PPD subjects, 73 PPWD and 74 HCs were included, and 68 metabolites were identified using GC-MS. The OPLS-DA model showed that there were 22 differential metabolites (14 upregulated and 8 downregulated) responsible for separating PPD subjects from HCs and PPWD. Meanwhile, a panel of five potential biomarkers - formate, succinate, 1-methylhistidine, α-glucose and dimethylamine - was identified. This panel could effectively distinguish PPD subjects from HCs and PPWD with an area under the curve (AUC) curve of 0.948 in the training set and 0.944 in the testing set. CONCLUSION These results demonstrated that the potential biomarker panel could aid in the future development of an objective diagnostic method for PPD.
Collapse
Affiliation(s)
- Lin Lin
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Shandong, People's Republic of China
| | - Xiao-Mei Chen
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Shandong, People's Republic of China
| | - Rong-Hua Liu
- Department of Obstetrics and Gynecology, Linyi People's Hospital, Shandong, People's Republic of China
| |
Collapse
|