201
|
Pope BS, Wood SK. Advances in understanding mechanisms and therapeutic targets to treat comorbid depression and cardiovascular disease. Neurosci Biobehav Rev 2020; 116:337-349. [PMID: 32598982 DOI: 10.1016/j.neubiorev.2020.06.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 06/16/2020] [Accepted: 06/24/2020] [Indexed: 02/07/2023]
Abstract
Chronic or repeated social stress exposure often precipitates the onset of depression and cardiovascular disease (CVD). Despite a clear clinical association between CVD and depression, the pathophysiology underlying these comorbid conditions is unclear. Chronic exposure to social stress can lead to immune system dysregulation, mitochondrial dysfunction, and vagal withdrawal. Further, regular physical exercise is well-known to exert cardioprotective effects, and accumulating evidence demonstrates the antidepressant effect of exercise. This review explores the contribution of inflammation, mitochondrial dysfunction, and vagal withdrawal to stress-induced depression and CVD. Evidence for therapeutic benefits of exercise, anti-inflammatory therapies, and vagus nerve stimulation are also reviewed. Benefits of targeted therapeutics of mitochondrial agents, anti-inflammatory therapies, and vagus nerve stimulation are discussed. Importantly, the ability of exercise to impact each of these factors is also reviewed. The current findings described here implicate a new direction for research, targeting the shared mechanisms underlying comorbid depression-CVD. This will guide the development of novel therapeutic strategies for the prevention and treatment of these stress-related pathologies, particularly within treatment-resistant populations.
Collapse
Affiliation(s)
- Brittany S Pope
- Department of Exercise Science, University of South Carolina Arnold School of Public Health, Columbia, SC, 20208, United States
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, 29209, United States; William Jennings Bryan Dorn Veterans Administration Medical Center, Columbia, SC, 29209, United States.
| |
Collapse
|
202
|
Yu YQ, Zhang YL, Wang Z, Liu Y, Li H, Zhou XF, Hu ZL, Dai RP. Involvement of proBDNF in Monocytes/Macrophages with Gastrointestinal Disorders in Depressive Mice. Neurotox Res 2020; 38:887-899. [PMID: 32588356 DOI: 10.1007/s12640-020-00235-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 05/19/2020] [Accepted: 06/07/2020] [Indexed: 12/19/2022]
Abstract
Major depressive disorders (MDD) are often comorbid with the gastrointestinal (GI) disorders. Brain-derived neurotrophic factor precursor (proBDNF) has been reported to contribute to the development of depression in mouse models. However, the role of proBDNF in depression-associated GI disorders is still unrevealed. Mice experienced unpredictable chronic mild stress (UCMS) procedure and were then intraperitoneally injected with fluoxetine (20 mg/kg). Open field test (OFT), forced swimming test (FST), and sucrose preference test (SPT) were performed to evaluate the severity of depression. Oral administration of food dye gel and histological staining were performed to assess GI transit and morphological alterations. QPCR was performed to assess the mRNA levels of inflammatory cytokines. Additionally, flow cytometry, immunohistochemistry, and immunofluorescence were performed to examine the expression and cellular localization of proBDNF. It was found that (a) in the peripheral blood, the expression of proBDNF and its receptor pan neurotrophin receptor 75 (p75NTR) in CD11b+ cells in depressive mice was higher than in controls; (b) the GI motility was decreased after the UCMS procedure and partly reversed by fluoxetine treatment; (c) proBDNF/p75NTR was highly expressed in macrophages in the intestinal lamina propria; (d) the upregulated proBDNF/p75NTR and the activated cytokines, including IL (interleukin)-1β, IL-6, IL-10, and IFN (interferon)-γ, were positively correlated with the depression and GI disorders, and were inhibited by fluoxetine treatment. UCMS procedure upregulated the expression of proBDNF and p75NTR in monocytes/macrophages of peripheral blood and intestinal lamina propria, which may be involved in the pathogenesis of depression-associated GI disorders. Fluoxetine reversed the GI dysfunction, infiltration of macrophages, and upregulation of proBDNF signaling in the depressive mice.
Collapse
Affiliation(s)
- Yun-Qing Yu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China.,Anesthesia Medical Research Center, Central South University, Changsha, China
| | - Yan-Ling Zhang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhe Wang
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu Liu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Hui Li
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China.,Anesthesia Medical Research Center, Central South University, Changsha, China
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Zhao-Lan Hu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Ru-Ping Dai
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China. .,Anesthesia Medical Research Center, Central South University, Changsha, China.
| |
Collapse
|
203
|
Abstract
The bone marrow (BM) is the primary site of postnatal hematopoiesis and hematopoietic stem cell (HSC) maintenance. The BM HSC niche is an essential microenvironment which evolves and responds to the physiological demands of HSCs. It is responsible for orchestrating the fate of HSCs and tightly regulates the processes that occur in the BM, including self-renewal, quiescence, engraftment, and lineage differentiation. However, the BM HSC niche is disturbed following hematological stress such as hematological malignancies, ionizing radiation, and chemotherapy, causing the cellular composition to alter and remodeling to occur. Consequently, hematopoietic recovery has been the focus of many recent studies and elucidating these mechanisms has great biological and clinical relevance, namely to exploit these mechanisms as a therapeutic treatment for hematopoietic malignancies and improve regeneration following BM injury. The sympathetic nervous system innervates the BM niche and regulates the migration of HSCs in and out of the BM under steady state. However, recent studies have investigated how sympathetic innervation and signaling are dysregulated under stress and the subsequent effect they have on hematopoiesis. Here, we provide an overview of distinct BM niches and how they contribute to HSC regulatory processes with a particular focus on neuronal regulation of HSCs under steady state and stress hematopoiesis.
Collapse
Affiliation(s)
- Claire Fielding
- Haematology, University of Cambridge, Cambridge, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- National Health Service Blood and Transplant, Cambridge, UK
| | - Simón Méndez-Ferrer
- Haematology, University of Cambridge, Cambridge, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- National Health Service Blood and Transplant, Cambridge, UK
| |
Collapse
|
204
|
Racial Discrimination, Inflammation, and Chronic Illness Among African American Women at Midlife: Support for the Weathering Perspective. J Racial Ethn Health Disparities 2020; 8:339-349. [PMID: 32488825 DOI: 10.1007/s40615-020-00786-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/14/2020] [Accepted: 05/26/2020] [Indexed: 12/16/2022]
Abstract
It is widely accepted that socioeconomic status (SES) is a fundamental cause of health inequality. There is evidence, however, that race is also a fundamental cause of disparities in health. Based on this idea, the weathering hypothesis developed by Geronimus and her colleagues views the elevated rates of illness and disability seen among Black Americans as a physiological response to the structural barriers, daily slights, and other threats to identity that comprise the Black experience. The current study tests the weathering hypothesis using chronic inflammation as an indicator of biological weathering. Specifically, we examine the extent to which persistent exposure to racial discrimination predicts elevated inflammation and, in turn, diagnosed chronic illness, after taking into account SES and several control variables. This mediation model was tested using zero-inflated Poisson path modeling with five waves of data collected from 391 African American women participating in the Family and Community Health Study (FACHS). A 13-item index was used to assess exposure to racial discrimination across 8 years. ELISA blood assays of seven cytokines central to the inflammatory response were used to construct an inflammatory index. Respondents reported their diagnosed chronic diseases. Consonant with the weathering hypothesis, persistent exposure to discrimination predicted inflammation which, in turn, predicted number of chronic diseases. This indirect effect was statistically significant. SES predicted having a chronic disease and the various controls showed no effect. The findings support the idea that race, like SES, is a fundamental cause of health inequalities.
Collapse
|
205
|
Frach L, Tierling S, Schwaiger M, Moser D, Heinrichs M, Hengstler JG, Walter J, Kumsta R. The mediating role of KITLG DNA methylation in the association between childhood adversity and cortisol stress reactivity does not replicate in monocytes. Psychoneuroendocrinology 2020; 116:104653. [PMID: 32203862 DOI: 10.1016/j.psyneuen.2020.104653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/19/2020] [Accepted: 03/09/2020] [Indexed: 01/01/2023]
Abstract
Adverse childhood experiences such as maltreatment or neglect are associated with mental health problems in adulthood. Changes in the regulation of the psychological and physiological stress reaction, mediated via epigenetic modifications, are discussed as potential mechanisms. This study aimed to replicate the role of DNA methylation of the KITLG gene in mediating the association between childhood adversity and stress-induced cortisol reactivity in a sample of adults reporting childhood adversity and a matched control group (N = 60). DNA was extracted from purified CD14+ monocytes and genome-wide DNA methylation was assessed with the 450k BeadChip for targeted replication and exploratory analyses. As previously reported, childhood adversity was associated with significantly lower cortisol reactivity to stress. We could neither replicate the association between KITLG DNA methylation and cortisol stress reactivity nor the association with childhood adversity. Moreover, DNA methylation of the target CpG (cg27512205) was not associated with KITLG mRNA expression in monocytes. Exploratory analyses of array-wide DNA methylation patterns showed no significant results for individual sites after correction for multiple testing - neither in association with childhood trauma nor with adult cortisol stress reactivity. The analysis of differentially methylated regions (DMRs) revealed two significant regions which both mapped to non-coding genes in the association with cortisol stress reactivity. The mediating role of DNA methylation of the KITLG locus in the association between childhood adversity and cortisol stress reactivity could not be replicated in monocytes. In addition to differences in investigated tissue, reasons for non-replication might include differences between samples in age, ethnicity, trauma severity, and cortisol reactivity.
Collapse
Affiliation(s)
- Leonard Frach
- Department of Genetic Psychology, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Sascha Tierling
- Department of Genetics/ Epigenetics, Saarland University, Saarbrücken, Germany
| | - Marion Schwaiger
- Department of Genetic Psychology, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Dirk Moser
- Department of Genetic Psychology, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Markus Heinrichs
- Department of Psychology, Laboratory for Biological and Personality Psychology, University of Freiburg, Freiburg, Germany; Freiburg Brain Imaging Center, University Medical Center, University of Freiburg, Freiburg, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), Dortmund, Germany
| | - Jörn Walter
- Department of Genetics/ Epigenetics, Saarland University, Saarbrücken, Germany
| | - Robert Kumsta
- Department of Genetic Psychology, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
206
|
Foertsch S, Reber SO. The role of physical trauma in social stress-induced immune activation. Neurosci Biobehav Rev 2020; 113:169-178. [DOI: 10.1016/j.neubiorev.2020.02.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 02/08/2020] [Accepted: 02/19/2020] [Indexed: 12/17/2022]
|
207
|
Rajalingam D, Nymoen I, Jacobsen DP, Eriksen MB, Dissen E, Nielsen MB, Einarsen SV, Gjerstad J. Repeated social defeat promotes persistent inflammatory changes in splenic myeloid cells; decreased expression of β-arrestin-2 (ARRB2) and increased expression of interleukin-6 (IL-6). BMC Neurosci 2020; 21:25. [PMID: 32471349 PMCID: PMC7260804 DOI: 10.1186/s12868-020-00574-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/20/2020] [Indexed: 12/16/2022] Open
Abstract
Background Previous studies suggest that persistent exposure to social stress in mammals may be associated with multiple physiological effects. Here, we examine the effects of social stress in rats, i.e. repeated social defeat, on behavior, hypothalamic–pituitary–adrenal (HPA)-axis and immune system. Methods A resident-intruder paradigm, where an intruder rat was exposed to social stress by a dominant resident rat for 1 hour each day for 7 consecutive days was used. The day after the last stress exposure in the paradigm the data were analyzed. Variation in social interaction was observed manually, whereas locomotion was analyzed off-line by a purpose-made software. Gene expression in the pituitary gland, adrenal gland and myeloid cells isolated from the spleen was measured by qPCR. Results The exposure to social stress induced decreased weight gain and increased locomotion. An increased nuclear receptor subfamily group C number 1 (NR3C1) expression in the pituitary gland was also shown. In myeloid cells harvested from the spleen, we observed decreased expression of the β2-adrenergic receptor (ADRB2) and β-arrestin-2 (ARRB2), but increased expression of interleukin-6 (IL-6). Subsequent analyses in the same cells showed that ARRB2 was negatively correlated with IL-6 following the stress exposure. Conclusion Our results show that that the experience of social stress in the form of repeated social defeat in rats is a potent stressor that in myeloid cells in the spleen promotes persistent inflammatory changes. Future research is needed to examine whether similar inflammatory changes also can explain the impact of social stress, such as bullying and harassment, among humans.
Collapse
Affiliation(s)
| | | | | | | | - Erik Dissen
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Morten Birkeland Nielsen
- Department of Psychosocial Science, University of Bergen, Bergen, Norway.,National Institute of Occupational Health, Oslo, Norway
| | | | - Johannes Gjerstad
- Department of Psychosocial Science, University of Bergen, Bergen, Norway.,National Institute of Occupational Health, Oslo, Norway
| |
Collapse
|
208
|
Sawicki CM, Humeidan ML, Sheridan JF. Neuroimmune Interactions in Pain and Stress: An Interdisciplinary Approach. Neuroscientist 2020; 27:113-128. [PMID: 32441204 DOI: 10.1177/1073858420914747] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mounting evidence indicates that disruptions in bidirectional communication pathways between the central nervous system (CNS) and peripheral immune system underlie the etiology of pathologic pain conditions. The purpose of this review is to focus on the cross-talk between these two systems in mediating nociceptive circuitry under various conditions, including nervous system disorders. Elevated and prolonged proinflammatory signaling in the CNS is argued to play a role in psychiatric illnesses and chronic pain states. Here we review current research on the dynamic interplay between altered nociceptive mechanisms, both peripheral and central, and physiological and behavioral changes associated with CNS disorders.
Collapse
Affiliation(s)
- Caroline M Sawicki
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Michelle L Humeidan
- Department of Anesthesiology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.,Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - John F Sheridan
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA.,Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.,Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, USA.,Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
209
|
|
210
|
Tawakol A, Osborne MT, Wang Y, Hammed B, Tung B, Patrich T, Oberfeld B, Ishai A, Shin LM, Nahrendorf M, Warner ET, Wasfy J, Fayad ZA, Koenen K, Ridker PM, Pitman RK, Armstrong KA. Stress-Associated Neurobiological Pathway Linking Socioeconomic Disparities to Cardiovascular Disease. J Am Coll Cardiol 2020; 73:3243-3255. [PMID: 31248544 DOI: 10.1016/j.jacc.2019.04.042] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/11/2019] [Accepted: 04/10/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND Lower socioeconomic status (SES) associates with a higher risk of major adverse cardiac events (MACE) via mechanisms that are not well understood. OBJECTIVES Because psychosocial stress is more prevalent among those with low SES, this study tested the hypothesis that stress-associated neurobiological pathways involving up-regulated inflammation in part mediate the link between lower SES and MACE. METHODS A total of 509 individuals, median age 55 years (interquartile range: 45 to 66 years), underwent clinically indicated whole-body 18F-fluorodeoxyglucose positron emission tomography/computed tomography imaging and met pre-defined inclusion criteria, including absence of known cardiovascular disease or active cancer. Baseline hematopoietic tissue activity, arterial inflammation, and in a subset of 289, resting amygdalar metabolism (a measure of stress-associated neural activity) were quantified using validated 18F-fluorodeoxyglucose positron emission tomography/computed tomography methods. SES was captured by neighborhood SES factors (e.g., median household income and crime). MACE within 5 years of imaging was adjudicated. RESULTS Over a median 4.0 years, 40 individuals experienced MACE. Baseline income inversely associated with amygdalar activity (standardized β: -0.157 [95% confidence interval (CI): -0.266 to -0.041]; p = 0.007) and arterial inflammation (β: -0.10 [95% CI: -0.18 to -0.14]; p = 0.022). Further, income associated with subsequent MACE (standardized hazard ratio: 0.67 [95% CI: 0.47 to 0.96]; p = 0.029) after multivariable adjustments. Mediation analysis demonstrated that the path of: ↓ neighborhood income to ↑ amygdalar activity to ↑ bone marrow activity to ↑ arterial inflammation to ↑ MACE was significant (β: -0.01 [95% CI: -0.06 to -0.001]; p < 0.05). CONCLUSIONS Lower SES: 1) associates with higher amygdalar activity; and 2) independently predicts MACE via a serial pathway that includes higher amygdalar activity, bone marrow activity, and arterial inflammation. These findings illuminate a stress-associated neurobiological mechanism by which SES disparities may potentiate adverse health outcomes.
Collapse
Affiliation(s)
- Ahmed Tawakol
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Cardiac MR PET CT Program, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| | - Michael T Osborne
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Cardiac MR PET CT Program, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ying Wang
- Cardiac MR PET CT Program, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Department of Nuclear Medicine, the First Hospital of China Medical University, Heping District, Shenyang, China
| | - Basma Hammed
- Cardiac MR PET CT Program, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Brian Tung
- Cardiac MR PET CT Program, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Tomas Patrich
- Cardiac MR PET CT Program, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Blake Oberfeld
- Cardiac MR PET CT Program, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Amorina Ishai
- Cardiac MR PET CT Program, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lisa M Shin
- Department of Psychology, Tufts University, Medford, Massachusetts
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Erica T Warner
- Clinical Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jason Wasfy
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Karestan Koenen
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Paul M Ridker
- Cardiology Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Roger K Pitman
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Katrina A Armstrong
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
211
|
Noren Hooten N, Evans MK. Age and poverty status alter the coding and noncoding transcriptome. Aging (Albany NY) 2020; 11:1189-1203. [PMID: 30779705 PMCID: PMC6402526 DOI: 10.18632/aging.101823] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/08/2019] [Indexed: 12/20/2022]
Abstract
Emerging evidence indicates that noncoding RNAs play regulatory roles in aging and disease. The functional roles of long noncoding RNAs (lncRNAs) in physiology and disease are not completely understood. Little is known about lncRNAs in the context of human aging and socio-environmental conditions. Microarray profiling of lncRNAs and mRNAs from peripheral blood mononuclear cells from young and old white (n=16) and African American (AA) males (n=16) living above or below poverty from the Healthy Aging in Neighborhoods of Diversity across the Life Span study revealed changes in both lncRNAs and mRNAs with age and poverty status in white males, but not in AA males. We validated lncRNA changes in an expanded cohort (n=40); CTD-3247F14.2, GAS5, H19, TERC and MEG3 changed significantly with age, whereas AK022914,GAS5, KB-1047C11.2, MEG3 and XLOC_003262 changed with poverty. Mitochondrial function and response to DNA damage and stress were pathways enriched in younger individuals. Response to stress, viral infection, and immune signals were pathways enriched in individuals living above poverty. These data show that both human age and a marker of social adversity influence lncRNA expression, which may provide insight about molecular pathways underlying aging and social factors that affect disparities in aging and disease.
Collapse
Affiliation(s)
- Nicole Noren Hooten
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
212
|
Mondelli V, Vernon AC. From early adversities to immune activation in psychiatric disorders: the role of the sympathetic nervous system. Clin Exp Immunol 2020; 197:319-328. [PMID: 31319436 DOI: 10.1111/cei.13351] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2019] [Indexed: 02/07/2023] Open
Abstract
Increased peripheral levels of cytokines and central microglial activation have been reported in patients with psychiatric disorders. The degree of both innate and adaptive immune activation is also associated with worse clinical outcomes and poor treatment response in these patients. Understanding the possible causes and mechanisms leading to this immune activation is therefore an important and necessary step for the development of novel and more effective treatment strategies for these patients. In this work, we review the evidence of literature pointing to childhood trauma as one of the main causes behind the increased immune activation in patients with psychiatric disorders. We then discuss the potential mechanisms linking the experience of early life adversity (ELA) to innate immune activation. Specifically, we focus on the innervation of the bone marrow from sympathetic nervous system (SNS) as a new and emerging mechanism that has the potential to bridge the observed increases in both central and peripheral inflammatory markers in patients exposed to ELA. Experimental studies in laboratory rodents suggest that SNS activation following early life stress exposure causes a shift in the profile of innate immune cells, with an increase in proinflammatory monocytes. In turn, these cells traffic to the brain and influence neural circuitry, which manifests as increased anxiety and other relevant behavioural phenotypes. To date, however, very few studies have been conducted to explore this candidate mechanism in humans. Future research is also needed to clarify whether these pathways could be partially reversible to improve prevention and treatment strategies in the future.
Collapse
Affiliation(s)
- V Mondelli
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Psychological Medicine, London, UK.,NIHR Biomedical Research Centre South London and Maudsley NHS Trust, London, UK
| | - A C Vernon
- King's College London, Institute of Psychiatry Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, London, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| |
Collapse
|
213
|
Zhang L, Pan J, Chen W, Jiang J, Huang J. Chronic stress-induced immune dysregulation in cancer: implications for initiation, progression, metastasis, and treatment. Am J Cancer Res 2020; 10:1294-1307. [PMID: 32509380 PMCID: PMC7269780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/02/2020] [Indexed: 06/11/2023] Open
Abstract
Psychological stress is a well-accepted risk factor in cancer initiation and progression. The explosive growth of psychoneuroimmunology research in the past decade has yielded an unprecedented wealth of information about the critical role of chronic stress in the immune dysfunction that influences tumor behaviors, which presents insights to mitigate distress and improve prognosis in cancer patients. Chronic stress exacerbates inflammation and causes a metabolism disorder, making it difficult for the organisms to maintain homeostasis and increasing its susceptibility to cancer. The shifted differentiation and redistribution of the immune system induced by chronic stress fail to combat cancer efficiently. Chronic stress increases the tumor-educated immune suppressive cells and impairs the cytotoxicity of cellular immunity, thereby promoting lymphatic metastasis and hematogenous metastasis. In addition, the efficacy of existing cancer therapies is undermined because chronic stress prevents the immune system from responding properly. Emerging stress-reduction measures have been administered to assist cancer patients to cope with the adverse effects of chronic stress. Here we systematically review the current molecular, cellular, physiological mechanisms about stress-mediated immune responses in the enhancement of tumor initiation and progression, remodeling of tumor microenvironment and impairment of anti-tumor treatment. We also summarize the potential clinically applicable stress-oriented strategies towards cancer and discuss briefly where important knowledge gaps remain.
Collapse
Affiliation(s)
- Leyi Zhang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310009, P. R. China
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province), Second Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310009, P. R. China
| | - Jun Pan
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310009, P. R. China
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province), Second Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310009, P. R. China
| | - Wuzhen Chen
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310009, P. R. China
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province), Second Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310009, P. R. China
| | - Jinxin Jiang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310009, P. R. China
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province), Second Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310009, P. R. China
| | - Jian Huang
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310009, P. R. China
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province), Second Affiliated Hospital, Zhejiang University School of MedicineHangzhou 310009, P. R. China
| |
Collapse
|
214
|
Community violence and cellular and cytokine indicators of inflammation in adolescents. Psychoneuroendocrinology 2020; 115:104628. [PMID: 32145570 PMCID: PMC7767083 DOI: 10.1016/j.psyneuen.2020.104628] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/14/2020] [Accepted: 02/13/2020] [Indexed: 01/14/2023]
Abstract
Neighborhood violence is associated with a range of health consequences but little is known about the biological processes involved. Research in disease pathogenesis has identified low-grade inflammation as a process that, beginning in the first decades of life, is both induced by chronic stress and a contributor to multiple cardiometabolic diseases that present throughout the lifecourse. Previous research has examined whether neighborhood violence is associated with inflammatory biomarkers, but has been limited to cytokine indicators of inflammation. In a sample of adolescents (n = 203) residing in Chicago, we tested cross-sectional associations between neighborhood violence and cellular and cytokine indicators of inflammation. Neighborhood-level violence was measured in multiple ways (as murder rates of Census block groups and as the sum of homicides within 1 and ½ mile zones) in the areas surrounding where youth lived and attended school. At the individual level, violence exposure was measured by self-report (direct victimization, witnessing violence, and/or victimization of family or friends in the past year). Adolescents residing in high-violence neighborhoods evidenced higher numbers of pro-inflammatory classical (CD14++CD16-) monocytes relative to those in less violent neighborhoods. In contrast, neighborhood-level violence was not consistently associated with cytokine levels across different model specifications. Self-reported violence exposure was also not consistently associated with inflammatory biomarkers. Neighborhood-level violence and self-reported violence exposure interacted, such that the positive association between neighborhood-level violence and classical monocytes was observed only among adolescents who reported being exposed to violence. Associations were largely specific to the neighborhoods in which youth lived as opposed to those in which they attended school. Findings provide the first evidence that youth residing in high-violence neighborhoods show mobilization of classical monocytes, suggesting a pro-inflammatory mechanism through which contextual stressors such as neighborhood violence may compromise health.
Collapse
|
215
|
Liang F, Yang S, Zhang Y, Hao T. Social housing promotes cognitive function through enhancing synaptic plasticity in APP/PS1 mice. Behav Brain Res 2020; 368:111910. [PMID: 31034995 DOI: 10.1016/j.bbr.2019.111910] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/06/2019] [Accepted: 04/12/2019] [Indexed: 12/11/2022]
Abstract
Previous studies have shown that loneliness increases the risk of AD (Alzheimer's disease) onset, while active and frequent social housing delays the onset of cognitive impairment. The mechanism of how this occurs remains unclear. In this study, we investigated how social interaction affected cognitive function and AD pathology in APP/PS1 (amyloid precursor protein/presenilin-1) mice. APP/PS1 mice were divided into either a social isolation (SI) group, a social contact with one mouse (SCO) group, or a social contact with five mice (SCF) group. Our results demonstrated that social housing improved the behavioral performance of APP/PS1 mice in Morris Water Maze testing, without significantly altering the rates of amyloid plaque deposition or amyloidogenic APP processes. Furthermore, the synaptic function, dendritic spine density, and complexity of neuronal network were notably increased in the SCF group, as compared to the SI and SCO groups. Additional protein and mRNA analyses of isolated astrocyte and microglia revealed that several glial genes related to regulation and anti-inflammatory progression were significantly upregulated, while pro-inflammatory markers were decreased. These findings highlight the important role of quality social communication (five mice not one mice) on maintaining neuronal function during AD pathogenesis and provide evidence to place great emphasis of family care of AD patients.
Collapse
Affiliation(s)
- Feiyu Liang
- Department of Geriatric Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shen Yang
- Departments of Neurology, Tai'an City Central Hospital, Tai'an, Shandong, 271000, China
| | - Yang Zhang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Tianpao Hao
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
216
|
Abstract
Unhealthy diet, lack of exercise, psychosocial stress, and insufficient sleep are increasingly prevalent modifiable risk factors for cardiovascular disease. Accumulating evidence indicates that these risk factors may fuel chronic inflammatory processes that are active in atherosclerosis and lead to myocardial infarction and stroke. In concert with hyperlipidemia, maladaptive immune system activities can contribute to disease progression and increase the probability of adverse events. In this review, we discuss recent insight into how the above modifiable risk factors influence innate immunity. Specifically, we focus on pathways that raise systemic myeloid cell numbers and modulate immune cell phenotypes, reviewing hematopoiesis, leukocyte trafficking, and innate immune cell accumulation in cardiovascular organs. Often, relevant mechanisms that begin with lifestyle choices and lead to cardiovascular events span multiple organ systems, including the central nervous, endocrine, metabolic, hematopoietic, immune and, finally, the cardiovascular system. We argue that deciphering such pathways provides not only support for preventive interventions but also opportunities to develop biomimetic immunomodulatory therapeutics that mitigate cardiovascular inflammation.
Collapse
Affiliation(s)
- Maximilian J Schloss
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston (M.J.S., F.K.S., M.N.).,Department of Radiology, Massachusetts General Hospital, Boston (M.J.S., F.K.S., M.N.)
| | - Filip K Swirski
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston (M.J.S., F.K.S., M.N.).,Department of Radiology, Massachusetts General Hospital, Boston (M.J.S., F.K.S., M.N.)
| | - Matthias Nahrendorf
- From the Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston (M.J.S., F.K.S., M.N.).,Department of Radiology, Massachusetts General Hospital, Boston (M.J.S., F.K.S., M.N.).,Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.N.).,Department of Internal Medicine I, University Hospital Wuerzburg, Germany (M.N.)
| |
Collapse
|
217
|
Motoyama S, Yamada H, Yamamoto K, Wakana N, Terada K, Kikai M, Wada N, Saburi M, Sugimoto T, Kubota H, Miyawaki D, Kami D, Ogata T, Ibi M, Yabe-Nishimura C, Matoba S. Social Stress Increases Vulnerability to High-Fat Diet-Induced Insulin Resistance by Enhancing Neutrophil Elastase Activity in Adipose Tissue. Cells 2020; 9:cells9040996. [PMID: 32316265 PMCID: PMC7226953 DOI: 10.3390/cells9040996] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/11/2020] [Accepted: 04/15/2020] [Indexed: 12/16/2022] Open
Abstract
Social stress (SS) has been linked to the development of cardiovascular disease (CVD), which is closely associated with insulin resistance (IR); however, the causal effect of SS on IR remains unclear. The 8-week-old male C57BL/6 mice were exposed to SS by housing with a larger CD-1 mouse in a shared home cage without physical contact for 10 consecutive days followed by high-fat diet (HFD) feeding. Control mice were housed in the same cage without a CD-1 mouse. After 6 weeks of HFD, insulin sensitivity was significantly impaired in stressed mice. While the percentage of classically activated macrophages in epididymal white adipose tissue (eWAT) was equivalent between the two groups, the percentage of lymphocyte antigen 6 complex locus G6D (Ly-6G)/neutrophil elastase (NE)-double positive cells markedly increased in stressed mice, accompanied by augmented NE activity assessed by ex vivo eWAT fluorescent imaging. Treatment with an NE inhibitor completely abrogated the insulin sensitivity impairment of stressed mice. In vitro NE release upon stimulation with a formyl peptide receptor 1 agonist was significantly higher in bone marrow neutrophils of stressed mice. Our findings show that SS-exposed mice are susceptible to the development of HFD-induced IR accompanied by augmented NE activity. Modulation of neutrophil function may represent a potential therapeutic target for SS-associated IR.
Collapse
Affiliation(s)
- Shinichiro Motoyama
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Hiroyuki Yamada
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Correspondence: ; Tel.: +81-75-251-5511
| | - Keita Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Noriyuki Wakana
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kensuke Terada
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Masakazu Kikai
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Naotoshi Wada
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Makoto Saburi
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Takeshi Sugimoto
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Hiroshi Kubota
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Daisuke Miyawaki
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Daisuke Kami
- Department of Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Takehiro Ogata
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Masakazu Ibi
- Department of Pharmacology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Chihiro Yabe-Nishimura
- Department of Pharmacology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
218
|
Kuhlman KR, Robles TF, Haydon MD, Dooley L, Boyle CC, Bower JE. Early life stress sensitizes individuals to the psychological correlates of mild fluctuations in inflammation. Dev Psychobiol 2020; 62:400-408. [PMID: 31489628 PMCID: PMC8211401 DOI: 10.1002/dev.21908] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 06/15/2019] [Accepted: 07/15/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Early life stress (ELS) has been linked to health disparities across the human lifespan, particularly increased risk for depression and its recurrence. In this study we explore two plausible and competing pathways through which ELS may lead to depression via inflammation. METHODS Participants (ages 18-22; n = 41) completed the Early Trauma Inventory as a measure of ELS. Participants then completed consecutive daily diaries of mood and other sickness behavior for the 7 days prior to and 7 days after receiving the annual influenza vaccine. Circulating concentrations of plasma interleukin-6 (IL-6) were measured immediately before and 24 hr after vaccination. RESULTS ELS was not associated with the magnitude of change in IL-6 from pre- to post-vaccine, however, exposure to ELS moderated the association between change in IL-6 from pre- to post-vaccine and changes in both cognitive difficulty and depressed mood. Individuals exposed to greater ELS showed greater psychological sensitivity to increases in IL-6. CONCLUSIONS Exposure to ELS may increase sensitivity to peripheral inflammation in the central nervous system. Future studies elaborating on the impact of ELS on the sensitivity of specific neural circuits and cells to inflammation are needed.
Collapse
Affiliation(s)
- Kate R. Kuhlman
- Department of Psychological Science, School of Social Ecology, University of California Irvine, Irvine, CA, USA
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Interdisciplinary Salivary Bioscience, University of California Irvine, Irvine, CA, USA
| | - Theodore F. Robles
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychology, University of California Los Angeles, Los Angeles, CA, USA
| | - Marcie D. Haydon
- Department of Psychological Science, School of Social Ecology, University of California Irvine, Irvine, CA, USA
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Chloe C. Boyle
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | - Julienne E. Bower
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychology, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
219
|
Population-based RNA profiling in Add Health finds social disparities in inflammatory and antiviral gene regulation to emerge by young adulthood. Proc Natl Acad Sci U S A 2020. [PMID: 32041883 DOI: 10.1073/pnas.1821367117/-/dcsupplemental] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Health in later life varies significantly by individual demographic characteristics such as age, sex, and race/ethnicity, as well as by social factors including socioeconomic status and geographic region. This study examined whether sociodemographic variations in the immune and inflammatory molecular underpinnings of chronic disease might emerge decades earlier in young adulthood. Using data from 1,069 young adults from the National Longitudinal Study of Adolescent to Adult Health (Add Health)-the largest nationally representative and ethnically diverse sample with peripheral blood transcriptome profiles-we analyzed variation in the expression of genes involved in inflammation and type I interferon (IFN) response as a function of individual demographic factors, sociodemographic conditions, and biobehavioral factors (smoking, drinking, and body mass index). Differential gene expression was most pronounced by sex, race/ethnicity, and body mass index (BMI), but transcriptome correlates were identified for every demographic dimension analyzed. Inflammation-related gene expression showed the most pronounced variation as a function of biobehavioral factors (BMI and smoking) whereas type I IFN-related transcripts varied most strongly as a function of individual demographic characteristics (sex and race/ethnicity). Bioinformatic analyses of transcription factor and immune-cell activation based on transcriptome-wide empirical differences identified additional effects of family poverty and geographic region. These results identify pervasive sociodemographic differences in immune-cell gene regulation that emerge by young adulthood and may help explain social disparities in the development of chronic illness and premature mortality at older ages.
Collapse
|
220
|
Cole SW, Shanahan MJ, Gaydosh L, Harris KM. Population-based RNA profiling in Add Health finds social disparities in inflammatory and antiviral gene regulation to emerge by young adulthood. Proc Natl Acad Sci U S A 2020; 117:4601-4608. [PMID: 32041883 PMCID: PMC7060722 DOI: 10.1073/pnas.1821367117] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Health in later life varies significantly by individual demographic characteristics such as age, sex, and race/ethnicity, as well as by social factors including socioeconomic status and geographic region. This study examined whether sociodemographic variations in the immune and inflammatory molecular underpinnings of chronic disease might emerge decades earlier in young adulthood. Using data from 1,069 young adults from the National Longitudinal Study of Adolescent to Adult Health (Add Health)-the largest nationally representative and ethnically diverse sample with peripheral blood transcriptome profiles-we analyzed variation in the expression of genes involved in inflammation and type I interferon (IFN) response as a function of individual demographic factors, sociodemographic conditions, and biobehavioral factors (smoking, drinking, and body mass index). Differential gene expression was most pronounced by sex, race/ethnicity, and body mass index (BMI), but transcriptome correlates were identified for every demographic dimension analyzed. Inflammation-related gene expression showed the most pronounced variation as a function of biobehavioral factors (BMI and smoking) whereas type I IFN-related transcripts varied most strongly as a function of individual demographic characteristics (sex and race/ethnicity). Bioinformatic analyses of transcription factor and immune-cell activation based on transcriptome-wide empirical differences identified additional effects of family poverty and geographic region. These results identify pervasive sociodemographic differences in immune-cell gene regulation that emerge by young adulthood and may help explain social disparities in the development of chronic illness and premature mortality at older ages.
Collapse
Affiliation(s)
- Steven W Cole
- School of Medicine, University of California, Los Angeles, CA 90095;
- Norman Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience & Human Behavior, University of California, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095
| | - Michael J Shanahan
- Department of Sociology, University of Zürich, CH 8050 Zürich, Switzerland
- Jacobs Center for Productive Youth Development, University of Zürich, CH 8050 Zürich, Switzerland
| | - Lauren Gaydosh
- Center for Medicine, Health, and Society, Vanderbilt University, Nashville, TN 37235
- Public Policy Studies, Department of Political Science, Vanderbilt University, Nashville, TN 37235
| | - Kathleen Mullan Harris
- Department of Sociology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516;
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516
| |
Collapse
|
221
|
Bazinet RP, Metherel AH, Chen CT, Shaikh SR, Nadjar A, Joffre C, Layé S. Brain eicosapentaenoic acid metabolism as a lead for novel therapeutics in major depression. Brain Behav Immun 2020; 85:21-28. [PMID: 31278982 DOI: 10.1016/j.bbi.2019.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
The results of several meta-analyses suggest that eicosapentaenoic acid (EPA) supplementation is therapeutic in managing the symptoms of major depression. It was previously assumed that because EPA is extremely low in the brain it did not cross the blood-brain barrier and any therapeutic effects it exerted would be via the periphery. However, more recent studies have established that EPA does enter the brain, but is rapidly metabolised following entry. While EPA does not accumulate within the brain, it is present in microglia and homeostatic mechanisms may regulate its esterification to phospholipids that serve important roles in cell signaling. Furthermore, a variety of signaling molecules from EPA have been described in the periphery and they have the potential to exert effects within the brain. If EPA is confirmed to be therapeutic in major depression as a result of adequately powered randomized clinical trials, future research on brain EPA metabolism could lead to the discovery of novel targets for treating or preventing major depression.
Collapse
Affiliation(s)
- Richard P Bazinet
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada.
| | - Adam H Metherel
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Chuck T Chen
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, North Bethesda, MD 20852, United States
| | - Saame Raza Shaikh
- Department of Nutrition, Gillings School of Global Public Health & School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| | - Agnes Nadjar
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| | - Corinne Joffre
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| | - Sophie Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Université de Bordeaux 2, 146 rue Léo Saignat, 33076 Bordeaux Cedex, France
| |
Collapse
|
222
|
Maturo MG, Soligo M, Gibson G, Manni L, Nardini C. The greater inflammatory pathway-high clinical potential by innovative predictive, preventive, and personalized medical approach. EPMA J 2020; 11:1-16. [PMID: 32140182 PMCID: PMC7028895 DOI: 10.1007/s13167-019-00195-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND LIMITATIONS Impaired wound healing (WH) and chronic inflammation are hallmarks of non-communicable diseases (NCDs). However, despite WH being a recognized player in NCDs, mainstream therapies focus on (un)targeted damping of the inflammatory response, leaving WH largely unaddressed, owing to three main factors. The first is the complexity of the pathway that links inflammation and wound healing; the second is the dual nature, local and systemic, of WH; and the third is the limited acknowledgement of genetic and contingent causes that disrupt physiologic progression of WH. PROPOSED APPROACH Here, in the frame of Predictive, Preventive, and Personalized Medicine (PPPM), we integrate and revisit current literature to offer a novel systemic view on the cues that can impact on the fate (acute or chronic inflammation) of WH, beyond the compartmentalization of medical disciplines and with the support of advanced computational biology. CONCLUSIONS This shall open to a broader understanding of the causes for WH going awry, offering new operational criteria for patients' stratification (prediction and personalization). While this may also offer improved options for targeted prevention, we will envisage new therapeutic strategies to reboot and/or boost WH, to enable its progression across its physiological phases, the first of which is a transient acute inflammatory response versus the chronic low-grade inflammation characteristic of NCDs.
Collapse
Affiliation(s)
- Maria Giovanna Maturo
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Greg Gibson
- Center for Integrative Genomics, School of Biological Sciences, Georgia Tech, Atlanta, GA USA
| | - Luigi Manni
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Christine Nardini
- IAC Institute for Applied Computing, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
- Bio Unit, Scientific and Medical Direction, SOL Group, Monza, Italy
| |
Collapse
|
223
|
Knight JM, Rizzo JD, Hari P, Pasquini MC, Giles KE, D'Souza A, Logan BR, Hamadani M, Chhabra S, Dhakal B, Shah N, Sriram D, Horowitz MM, Cole SW. Propranolol inhibits molecular risk markers in HCT recipients: a phase 2 randomized controlled biomarker trial. Blood Adv 2020; 4:467-476. [PMID: 32027744 PMCID: PMC7013267 DOI: 10.1182/bloodadvances.2019000765] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/11/2019] [Indexed: 12/26/2022] Open
Abstract
Preclinical research shows that stress-induced activation of the sympathetic nervous system can promote hematopoietic malignancies via β-adrenoreceptor-mediated molecular pathways. Hematopoietic cell transplant (HCT) recipients exposed to conditions of chronic stress show activation of a conserved transcriptional response to adversity (CTRA) gene expression profile, which in turn is associated with increased relapse and decreased disease-free survival. We conducted a randomized controlled phase 2 biomarker trial testing the impact of the nonselective β-antagonist propranolol on CTRA-related gene expression of 25 individuals receiving an autologous HCT for multiple myeloma. Propranolol was administered for 1 week prior to and 4 weeks following HCT. Blood was collected at baseline, day -2, and day +28. Intention-to-treat analyses controlling for demographic characteristics, high-risk disease (International Myeloma Working Group risk score), and tumor stage tested effects on a 53-gene CTRA indicator profile and measures of CTRA-related cellular processes in peripheral blood mononuclear cells. Twelve participants were randomized to the intervention and 13 to the control. Relative to the control group, propranolol-treated patients showed greater decreases from baseline to HCT day -2 and day +28 for both CTRA gene expression (P = .017) and bioinformatic measures of CD16- classical monocyte activation (P = .005). Propranolol-treated patients also showed relative upregulation of CD34+ cell-associated gene transcripts (P = .011) and relative downregulation of myeloid progenitor-containing CD33+ cell-associated gene transcripts (P = .001). Ancillary analyses identified nonsignificant trends toward accelerated engraftment and reduced posttransplant infections in propranolol-treated patients. Peri-HCT propranolol inhibits cellular and molecular pathways associated with adverse outcomes. Changes in these pathways make propranolol a potential candidate for adjunctive therapy in cancer-related HCT.
Collapse
Affiliation(s)
- Jennifer M Knight
- Department of Psychiatry
- Department of Medicine
- Department of Microbiology & Immunology
- Section of Blood and Marrow Transplantation & Cellular Therapy, Division of Hematology/Oncology, Department of Medicine, and
| | - J Douglas Rizzo
- Section of Blood and Marrow Transplantation & Cellular Therapy, Division of Hematology/Oncology, Department of Medicine, and
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI
| | - Parameswaran Hari
- Section of Blood and Marrow Transplantation & Cellular Therapy, Division of Hematology/Oncology, Department of Medicine, and
| | - Marcelo C Pasquini
- Section of Blood and Marrow Transplantation & Cellular Therapy, Division of Hematology/Oncology, Department of Medicine, and
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI
| | | | - Anita D'Souza
- Section of Blood and Marrow Transplantation & Cellular Therapy, Division of Hematology/Oncology, Department of Medicine, and
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI
| | - Brent R Logan
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI
- Division of Biostatistics, Institute for Health & Society, Medical College of Wisconsin, Milwaukee, WI; and
| | - Mehdi Hamadani
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI
- Division of Biostatistics, Institute for Health & Society, Medical College of Wisconsin, Milwaukee, WI; and
| | - Saurabh Chhabra
- Section of Blood and Marrow Transplantation & Cellular Therapy, Division of Hematology/Oncology, Department of Medicine, and
| | - Binod Dhakal
- Section of Blood and Marrow Transplantation & Cellular Therapy, Division of Hematology/Oncology, Department of Medicine, and
| | - Nirav Shah
- Section of Blood and Marrow Transplantation & Cellular Therapy, Division of Hematology/Oncology, Department of Medicine, and
| | - Deepika Sriram
- Section of Blood and Marrow Transplantation & Cellular Therapy, Division of Hematology/Oncology, Department of Medicine, and
| | - Mary M Horowitz
- Section of Blood and Marrow Transplantation & Cellular Therapy, Division of Hematology/Oncology, Department of Medicine, and
- Center for International Blood and Marrow Transplant Research, Medical College of Wisconsin, Milwaukee, WI
| | - Steve W Cole
- Division of Hematology-Oncology, Department of Medicine, and
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
224
|
Munshi S, Loh MK, Ferrara N, DeJoseph MR, Ritger A, Padival M, Record MJ, Urban JH, Rosenkranz JA. Repeated stress induces a pro-inflammatory state, increases amygdala neuronal and microglial activation, and causes anxiety in adult male rats. Brain Behav Immun 2020; 84:180-199. [PMID: 31785394 PMCID: PMC7010555 DOI: 10.1016/j.bbi.2019.11.023] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/25/2022] Open
Abstract
A link exists between immune function and psychiatric conditions, particularly depressive and anxiety disorders. Psychological stress is a powerful trigger for these disorders and stress influences immune state. However, the nature of peripheral immune changes after stress conflicts across studies, perhaps due to the focus on few measures of pro-inflammatory or anti-inflammatory processes. The basolateral amygdala (BLA) is critical for emotion, and plays an important role in the effects of stress on anxiety. As such, it may be a primary central nervous system (CNS) mediator for the effects of peripheral immune changes on anxiety after stress. Therefore, this study aimed to delineate the influence of stress on peripheral pro-inflammatory and anti-inflammatory aspects, BLA immune activation, and its impact on BLA neuronal activity. To produce a more encompassing view of peripheral immune changes, this study used a less restrictive approach to categorize and group peripheral immune changes. We found that repeated social defeat stress in adult male Sprague-Dawley rats increased the frequencies of mature T-cells positive for intracellular type 2-like cytokine and serum pro-inflammatory cytokines. Principal component analysis and hierarchical clustering was used to guide grouping of T-cells and cytokines, producing unique profiles. Stress shifted the balance towards a specific set that included mostly type 2-like T-cells and pro-inflammatory cytokines. Within the CNS component, repeated stress caused an increase of activated microglia in the BLA, increased anxiety-like behaviors across several assays, and increased BLA neuronal firing in vivo that was prevented by blockade of microglia activation. Because repeated stress can trigger anxiety states by actions in the BLA, and altered immune function can trigger anxiety, these results suggest that repeated stress may trigger anxiety-like behaviors by inducing a pro-inflammatory state in the periphery and the BLA. These results begin to uncover how stress may recruit the immune system to alter the function of brain regions critical to emotion.
Collapse
Affiliation(s)
- Soumyabrata Munshi
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Department of Foundational Sciences and Humanities, Neuroscience, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Maxine K. Loh
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Nicole Ferrara
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - M. Regina DeJoseph
- Department of Foundational Sciences and Humanities, Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Alexandra Ritger
- Department of Foundational Sciences and Humanities, Neuroscience, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Mallika Padival
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Matthew J. Record
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Janice H. Urban
- Department of Foundational Sciences and Humanities, Physiology and Biophysics, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - J. Amiel Rosenkranz
- Department of Foundational Sciences and Humanities, Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.,Corresponding Author: J. Amiel Rosenkranz, Ph.D., Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA., Telephone: 847-578-8680; Fax: 847-578-3268,
| |
Collapse
|
225
|
Agrawal P, Kao D, Chung P, Looger LL. The neuropeptide Drosulfakinin regulates social isolation-induced aggression in Drosophila. J Exp Biol 2020; 223:jeb207407. [PMID: 31900346 PMCID: PMC7033730 DOI: 10.1242/jeb.207407] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 12/19/2019] [Indexed: 01/09/2023]
Abstract
Social isolation strongly modulates behavior across the animal kingdom. We utilized the fruit fly Drosophila melanogaster to study social isolation-driven changes in animal behavior and gene expression in the brain. RNA-seq identified several head-expressed genes strongly responding to social isolation or enrichment. Of particular interest, social isolation downregulated expression of the gene encoding the neuropeptide Drosulfakinin (Dsk), the homologue of vertebrate cholecystokinin (CCK), which is critical for many mammalian social behaviors. Dsk knockdown significantly increased social isolation-induced aggression. Genetic activation or silencing of Dsk neurons each similarly increased isolation-driven aggression. Our results suggest a U-shaped dependence of social isolation-induced aggressive behavior on Dsk signaling, similar to the actions of many neuromodulators in other contexts.
Collapse
Affiliation(s)
- Pavan Agrawal
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Damian Kao
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Phuong Chung
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Loren L Looger
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| |
Collapse
|
226
|
Dieckmann L, Cole S, Kumsta R. Stress genomics revisited: gene co-expression analysis identifies molecular signatures associated with childhood adversity. Transl Psychiatry 2020; 10:34. [PMID: 32066736 PMCID: PMC7026041 DOI: 10.1038/s41398-020-0730-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/19/2019] [Accepted: 01/08/2020] [Indexed: 12/15/2022] Open
Abstract
Childhood adversity is related to an increased risk for psychopathology in adulthood. Altered regulation of stress response systems, as well as the changes in stress-immune interplay have been suggested as potential mechanisms underlying these long-term effects. We have previously shown altered transcriptional responses to acute psychosocial stress in adults reporting the experience of childhood adversity. Here, we extend these analyses using a network approach. We performed a co-expression network analysis of genome-wide mRNA data derived from isolated monocytes, sampled 3 h after stress exposure from healthy adults, who experienced childhood adversity and a matched control group without adverse childhood experiences. Thirteen co-expression modules were identified, of which four modules were enriched for genes related to immune system function. Gene set enrichment analysis showed differential module activity between the early adversity and control group. In line with previous findings reporting a pro-inflammatory bias following childhood adversity, one module included genes associated with pro-inflammatory function (hub genes: IL6, TM4SF1, ADAMTS4, CYR61, CCDC3), more strongly expressed in the early adversity group. Another module downregulated in the early adversity group was related to platelet activation and wound healing (hub genes: GP9, CMTM5, TUBB1, GNG11, PF4), and resembled a co-expression module previously found over-expressed in post-traumatic stress disorder resilient soldiers. These discovery analysis results provide a system wide and more holistic understanding of gene expression programs associated with childhood adversity. Furthermore, identified hub genes can be used in directed hypothesis testing in future studies.
Collapse
Affiliation(s)
- Linda Dieckmann
- grid.5570.70000 0004 0490 981XDepartment of Genetic Psychology, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Steve Cole
- grid.19006.3e0000 0000 9632 6718Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Cousins Center for Psychoneuroimmunology, University of California, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA USA
| | - Robert Kumsta
- Department of Genetic Psychology, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
227
|
Li MJ, Takada S, Okafor CN, Gorbach PM, Shoptaw SJ, Cole SW. Experienced homophobia and gene expression alterations in Black and Latino men who have sex with men in Los Angeles County. Brain Behav Immun 2020; 83:120-125. [PMID: 31563693 PMCID: PMC6906252 DOI: 10.1016/j.bbi.2019.09.021] [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: 06/28/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 01/17/2023] Open
Abstract
Men who have sex with men (MSM) experience high rates of homophobic victimization, which is linked to myriad chronic physical and mental health disparities. Social adversity such as rejection, isolation, and racial discrimination can induce a conserved transcriptional response to adversity (CTRA) involving upregulation of proinflammatory genes and downregulation of type I interferon and antibody synthesis genes. This study specifically examines whether homophobic victimization is associated with expression of CTRA profiles in Black and Latino MSM living in Los Angeles. Analyses linked behavioral survey data with quantified RNA from leukocytes from blood samples of 70 participants over 12 months. CTRA gene expression was increased by 3.1-fold in MSM who experienced homophobic victimization while adjusting for major leukocyte subsets and sociodemographics. Accounting for all these factors, CTRA gene expression was significantly enhanced in MSM who identified as Black compared to Latino. Our findings identify experiences of homophobic victimization as drivers of inflammatory and type I interferon gene expression profiles, which can contribute to physical and mental health challenges in Black and Latino MSM.
Collapse
Affiliation(s)
- Michael J. Li
- Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles,Center for Behavioral and Addiction Medicine, Department of Family Medicine, David Geffen School of Medicine, University of California, Los Angeles,Department of Family Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Sae Takada
- National Clinician Scholars Program UCLA, Division of General Internal Medicine and Health Services Research, Department of Medicine, Los Angeles, CA,Veterans Affairs, Health Services Research & Development, Center for the Study of Healthcare Innovation, Implementation, & Policy, Los Angeles, CA
| | - Chukwuemeka N. Okafor
- Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles,Center for Behavioral and Addiction Medicine, Department of Family Medicine, David Geffen School of Medicine, University of California, Los Angeles,Department of Family Medicine, David Geffen School of Medicine, University of California, Los Angeles
| | - Pamina M. Gorbach
- Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles,Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles
| | - Steven J. Shoptaw
- Center for Behavioral and Addiction Medicine, Department of Family Medicine, David Geffen School of Medicine, University of California, Los Angeles,Department of Family Medicine, David Geffen School of Medicine, University of California, Los Angeles,Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles
| | - Steven W. Cole
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles,Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles
| |
Collapse
|
228
|
Seeman T, Merkin SS, Goldwater D, Cole SW. Intergenerational mentoring, eudaimonic well-being and gene regulation in older adults: A pilot study. Psychoneuroendocrinology 2020; 111:104468. [PMID: 31589939 DOI: 10.1016/j.psyneuen.2019.104468] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 12/29/2022]
Abstract
To clarify the biological mechanisms underlying the relationship between pro-social behavior and health, this pilot study examined the impact of a 9-month intergenerational helping intervention on conserved transcriptional response to adversity (CTRA) gene expression profiles, which are characterized by up-regulation of genes involved in inflammation and down-regulation of genes involved in antiviral defenses. The Generation Xchange program trains and places older (age 50+) volunteers in K-3rd grade classrooms to aid students' academic development (reading and math) and address behavioral issues (e.g., inability to focus during class, behaviors that disrupt class). Volunteers were predominately women (89%) and African American (94%) from the neighborhoods around the schools. Repeated measures planned contrast analysis of 53 CTRA indicator transcripts in 50 blood samples collected from 18 individuals on 2-3 occasions revealed a significant reduction in CTRA gene expression from baseline to the average of 3- and 9-month follow-up. The magnitude of individual decrease in CTRA gene expression correlated with the magnitude of individual increase in eudaimonic well-being over time (net of changes in hedonic well-being). In addition to clarifying biological pathways through which pro-social behavior might impact health, these pilot data suggest that the GenX program may have favorable effects on immune cell gene regulation.
Collapse
Affiliation(s)
- Teresa Seeman
- Division of Geriatrics, David Geffen School of Medicine at UCLA, Le Conte Ave, Suite 2339, Los Angeles, CA, 900945-1687, United States.
| | - Sharon Stein Merkin
- Division of Geriatrics, David Geffen School of Medicine at UCLA, Le Conte Ave, Suite 2339, Los Angeles, CA, 900945-1687, United States.
| | - Deena Goldwater
- Division of Geriatrics, David Geffen School of Medicine at UCLA, Le Conte Ave, Suite 2339, Los Angeles, CA, 900945-1687, United States.
| | - Steven W Cole
- Division of Hematology-Oncology, Department of Medicine, Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, 11-934 Factor Bldg, Los Angeles, CA, 90095-1678, United States
| |
Collapse
|
229
|
McKim DB, Yin W, Wang Y, Cole SW, Godbout JP, Sheridan JF. Social Stress Mobilizes Hematopoietic Stem Cells to Establish Persistent Splenic Myelopoiesis. Cell Rep 2019; 25:2552-2562.e3. [PMID: 30485819 DOI: 10.1016/j.celrep.2018.10.102] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/10/2018] [Accepted: 10/26/2018] [Indexed: 01/09/2023] Open
Abstract
Psychosocial stress accelerates myelopoietic production of monocytes and neutrophils that contributes to a variety of health complications ranging from atherosclerosis to anxiety. Here, we show that social stress in mice mobilizes hematopoietic stem progenitor cells (HSPCs) from the bone marrow that enter circulation, engraft into the spleen, and establish a persistent extramedullary hematopoietic depot. These splenic progenitors actively proliferate and differentiate into multiple cell types, including monocytes, neutrophils, and erythrocytes. Splenic erythropoiesis partially abrogates stress-induced anemia. Repeated injection with isoprenaline induces progenitor mobilization to the spleen, identifying a key role for β-adrenergic signaling. Moreover, protracted splenic production of CD11b+ cells persists for at least 24 days after the cessation of social stress. Thus, chronic stress establishes a persistent extramedullary hematopoietic depot that can modify a wide range of chronic disease processes and alter homeostasis of the bi-directional regulatory axis between the nervous and immune systems.
Collapse
Affiliation(s)
- Daniel B McKim
- Division of Biosciences, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Wenyuan Yin
- Division of Biosciences, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Yufen Wang
- Division of Biosciences, The Ohio State University, Columbus, OH, USA
| | - Steve W Cole
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA
| | - John F Sheridan
- Division of Biosciences, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University, Columbus, OH, USA; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA; Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
230
|
Nagata JM, Palar K, Gooding HC, Garber AK, Bibbins-Domingo K, Weiser SD. Food Insecurity and Chronic Disease in US Young Adults: Findings from the National Longitudinal Study of Adolescent to Adult Health. J Gen Intern Med 2019; 34:2756-2762. [PMID: 31576509 PMCID: PMC6854148 DOI: 10.1007/s11606-019-05317-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 06/07/2019] [Accepted: 07/23/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND Food insecurity, or the limited or uncertain access to food resulting from inadequate financial resources, is associated with a higher prevalence of chronic disease in adulthood. Little is known about these relationships specifically in young adulthood, an important time for the development of chronic disease. OBJECTIVE To determine the association between food insecurity and chronic disease including diabetes, hypertension, obesity, and obstructive airway disease in a nationally representative sample of US young adults. DESIGN Cross-sectional nationally representative data collected from Wave IV (2008) of the National Longitudinal Study of Adolescent to Adult Health was analyzed using multiple logistic regression models. PARTICIPANTS US young adults ages 24-32 years old MAIN MEASURES: Food insecurity and general health; self-reported diabetes, hypertension, hyperlipidemia, "very overweight," and obstructive airway disease; measured obesity derived from body mass index; and inadequate disease control (hemoglobin A1c ≥ 7.0%, blood pressure ≥ 140/90 mmHg) among those with reported diabetes and hypertension. KEY RESULTS Of the 14,786 young adults in the sample, 11% were food insecure. Food-insecure young adults had greater odds of self-reported poor health (2.63, 95% confidence interval (CI) 1.63-4.24), diabetes (1.67, 95% CI 1.18-2.37), hypertension (1.40, 95% CI 1.14-1.72), being "very overweight" (1.30, 95% CI 1.08-1.57), and obstructive airway disease (1.48, 95% CI 1.22-1.80) in adjusted models compared with young adults who were food secure. Food insecurity was not associated with inadequate disease control among those with diabetes or hypertension. CONCLUSIONS Food insecurity is associated with several self-reported chronic diseases and obesity in young adulthood. Health care providers should screen for food insecurity in young adults and provide referrals when appropriate. Future research should evaluate the impact of early interventions to combat food insecurity on the prevention of downstream health effects in later adulthood.
Collapse
Affiliation(s)
- Jason M Nagata
- Department of Pediatrics, University of California, San Francisco, 550 16th Street, 4th Floor, Box 0110, San Francisco, CA, 94158, USA.
| | - Kartika Palar
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Holly C Gooding
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Andrea K Garber
- Department of Pediatrics, University of California, San Francisco, 550 16th Street, 4th Floor, Box 0110, San Francisco, CA, 94158, USA
| | - Kirsten Bibbins-Domingo
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Sheri D Weiser
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
231
|
Hiller JG, Cole SW, Crone EM, Byrne DJ, Shackleford DM, Pang JMB, Henderson MA, Nightingale SS, Ho KM, Myles PS, Fox S, Riedel B, Sloan EK. Preoperative β-Blockade with Propranolol Reduces Biomarkers of Metastasis in Breast Cancer: A Phase II Randomized Trial. Clin Cancer Res 2019; 26:1803-1811. [PMID: 31754048 DOI: 10.1158/1078-0432.ccr-19-2641] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/01/2019] [Accepted: 11/18/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE The majority of deaths from breast cancer occur following the development of metastatic disease, a process inhibited by β-blockers in preclinical studies. This phase II randomized controlled trial evaluated the effect of preoperative β-blockade with propranolol on biomarkers of metastatic potential and the immune cell profile within the primary tumor of patients with breast cancer. PATIENTS AND METHODS In this triple-blind placebo-controlled clinical trial, 60 patients were randomly assigned to receive an escalating dose of oral propranolol (n = 30; 80-160 mg daily) or placebo (n = 30) for 7 days prior to surgery. The primary endpoint investigated the effect of propranolol on prometastatic and proinflammatory gene expression within the primary tumor. RESULTS Propranolol downregulated primary tumor expression of mesenchymal genes (P = 0.002) without affecting epithelial gene expression (P = 0.21). Bioinformatic analyses implicated downregulation of Snail/Slug (P = 0.03), NF-κB/Rel (P < 0.01), and AP-1 (P < 0.01) transcription factors in structuring the observed transcriptome alterations, and identified changes in intratumoral neutrophil, natural killer cell, and dendritic cell recruitment (all P < 0.01). Patients with clinical evidence of drug response (lowered heart rate and blood pressure) demonstrated elevated tumor infiltration of CD68+ macrophages and CD8+ T cells. CONCLUSIONS One week of β-blockade with propranolol reduced intratumoral mesenchymal polarization and promoted immune cell infiltration in early-stage surgically-resectable breast cancer. These results show that β-blockade reduces biomarkers associated with metastatic potential, and support the need for larger phase III clinical trials powered to detect the impact of β-blockade on cancer recurrence and survival.See related commentary by Blaes et al., p. 1781.
Collapse
Affiliation(s)
- Jonathan G Hiller
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, Melbourne, Australia. .,Faculty of Medicine, Dentistry and Health Sciences, Centre for Integrated Critical Care Medicine, University of Melbourne, Parkville, Melbourne, Australia.,Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Australia.,Department of Anaesthesia and Perioperative Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Steven W Cole
- Department of Medicine, Division of Hematology-Oncology, UCLA Molecular Biology Institute, University of California Los Angeles, Los Angeles, California.,Department of Psychiatry and Semel Institute for Neuroscience and Human Behavior, Jonsson Comprehensive Cancer Center, UCLA Molecular Biology Institute, University of California Los Angeles, Los Angeles, California.,UCLA AIDS Institute, University of California Los Angeles, Los Angeles, California
| | - Elizabeth M Crone
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, Melbourne, Australia
| | - David J Byrne
- Department of Pathology, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, Melbourne, Australia
| | - David M Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Australia
| | - Jia-Min B Pang
- Department of Pathology, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, Melbourne, Australia
| | - Michael A Henderson
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, Melbourne, Australia
| | - Sophie S Nightingale
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, Melbourne, Australia
| | - Kwok M Ho
- Department of Intensive Care Medicine, Royal Perth Hospital, Perth, Australia
| | - Paul S Myles
- Department of Anaesthesia and Perioperative Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Stephen Fox
- Department of Pathology, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, Melbourne, Australia
| | - Bernhard Riedel
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, Melbourne, Australia.,Faculty of Medicine, Dentistry and Health Sciences, Centre for Integrated Critical Care Medicine, University of Melbourne, Parkville, Melbourne, Australia.,Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Australia.,The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Erica K Sloan
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, Melbourne, Australia. .,Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Australia.,Department of Medicine, Division of Hematology-Oncology, UCLA Molecular Biology Institute, University of California Los Angeles, Los Angeles, California.,Department of Psychiatry and Semel Institute for Neuroscience and Human Behavior, Jonsson Comprehensive Cancer Center, UCLA Molecular Biology Institute, University of California Los Angeles, Los Angeles, California.,UCLA AIDS Institute, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
232
|
Holmes L, Shutman E, Chinaka C, Deepika K, Pelaez L, Dabney KW. Aberrant Epigenomic Modulation of Glucocorticoid Receptor Gene (NR3C1) in Early Life Stress and Major Depressive Disorder Correlation: Systematic Review and Quantitative Evidence Synthesis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16214280. [PMID: 31689998 PMCID: PMC6861987 DOI: 10.3390/ijerph16214280] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/27/2019] [Accepted: 10/28/2019] [Indexed: 12/21/2022]
Abstract
Early life stress (ELS) induced by psychological trauma, child maltreatment, maternal separation, and domestic violence predisposes to psycho-behavioral pathologies during adulthood, namely major depressive disorder (MDD), anxiety, and bipolar affective disorder. While environmental data are available in illustrating this association, data remain to be established on the epigenomic underpinning of the nexus between ELS and MDD predisposition. Specifically, despite the observed aberrant epigenomic modulation of the NR3C1, a glucocorticoid receptor gene, in early social adversity and social threats in animal and human models, reliable scientific data for intervention mapping in reducing social adversity and improving human health is required. We sought to synthesize the findings of studies evaluating (a) epigenomic modulations, mainly DNA methylation resulting in MDD following ELS, (b) epigenomic modifications associated with ELS, and (c) epigenomic alterations associated with MDD. A systematic review and quantitative evidence synthesis (QES) were utilized with the random effect meta-analytic procedure. The search strategy involved both the PubMed and hand search of relevant references. Of the 1534 studies identified through electronic search, 592 studies were screened, 11 met the eligibility criteria for inclusion in the QES, and 5 examined ELS and MDD; 4 studies assessed epigenomic modulation and ELS, while 2 studies examined epigenomic modulations and MDD. The dense DNA methylation of the 1F exon of the NR3C1, implying the hypermethylated region of the glucocorticoid receptor gene, was observed in the nexus between ELS and MDD, common effect size (CES) = 14.96, 95%CI, 10.06-19.85. With respect to epigenomic modulation associated with child ELS, hypermethylation was observed, CES = 23.2%, 95%CI, 8.00-38.48. In addition, marginal epigenomic alteration was indicated in MDD, where hypermethylation was associated with increased risk of MDD, CES = 2.12%, 95%CI, -0.63-4.86. Substantial evidence supports the implication of NR3C1 and environmental interaction, mainly DNA methylation, in the predisposition to MDD following ELS. This QES further supports aberrant epigenomic modulation identified in ELS as well as major depressive episodes involving dysfunctional glucocorticoid-mediated negative feedback as a result of allostatic overload. These findings recommend prospective investigation of social adversity and its predisposition to the MDD epidemic via aberrant epigenomic modulation. Such data will facilitate early intervention mapping in reducing MDD in the United States population.
Collapse
Affiliation(s)
- Laurens Holmes
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
- Biological Sciences Department, University of Delaware, Newark, DE 19716, USA.
- College of Population Health, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Emily Shutman
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
- Biological Sciences Department, Haverford College, Haverford, PA 19041, USA.
| | - Chinacherem Chinaka
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
- Department of public health, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
- Community Environmental Health Department, Old Dominion University, Norfolk, VA 23507, USA.
| | - Kerti Deepika
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
| | - Lavisha Pelaez
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
| | - Kirk W Dabney
- Nemours Healthcare System for Children, Translational Health Disparities Science Research Program, Wilmington, DE 19803, USA.
- Sidney Kimmel Medical School, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
233
|
Snodgrass JG, Lacy MG, Dengah HJF, Polzer ER, Else RJ, Arevalo JMG, Cole SW. Positive mental well-being and immune transcriptional profiles in highly involved videogame players. Brain Behav Immun 2019; 82:84-92. [PMID: 31376495 PMCID: PMC6800642 DOI: 10.1016/j.bbi.2019.07.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 12/14/2022] Open
Abstract
Previous research has identified a link between experiencing life as meaningful and purposeful-what is referred to as "eudaimonia"-and reduced expression of a stress-induced gene profile known as the "conserved transcriptional response to adversity" (CTRA). In the current study, we examine whether similar links between eudaimonic well-being and CTRA reduction occur in a sample of 56 individuals with a particularly strong engagement with virtual worlds: avid online videogame players. Results consistently linked higher eudaimonic well-being, and more specifically the social well-being subdomain of eudaimonia, to lower levels of CTRA gene expression. That favorable psychobiological relationship between eudaimonia and CTRA appeared most strongly among individuals reporting high levels of positive psychosocial involvement with gaming. Findings are consistent with the hypothesis that committed social/recreational activity may help damp CTRA expression especially among persons who are already experiencing some kind of threshold of positive eudaimonic experience.
Collapse
Affiliation(s)
- Jeffrey G Snodgrass
- Department of Anthropology and Geography, Colorado State University, Fort Collins, CO 80523-1787, USA.
| | - Michael G Lacy
- Department of Sociology, Colorado State University, Fort Collins, CO 80523-1784, USA
| | - H J François Dengah
- Department of Sociology, Social Work, and Anthropology, Utah State University, Logan, UT 84322-0230, USA
| | - Evan R Polzer
- Department of Emergency Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Robert J Else
- Department of Anthropology, University of Alabama, Tuscaloosa, AL 35487-0210, USA
| | - Jesusa M G Arevalo
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles School of Medicine, Los Angeles, CA 90095, USA
| | - Steven W Cole
- Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles School of Medicine, Los Angeles, CA 90095, USA; Department of Psychiatry & Biobehavioral Sciences, University of California, Los Angeles School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
234
|
Mesenchymal Stromal Cells Modulate Peripheral Stress-Induced Innate Immune Activation Indirectly Limiting the Emergence of Neuroinflammation-Driven Depressive and Anxiety-like Behaviors. Biol Psychiatry 2019; 86:712-724. [PMID: 31521333 DOI: 10.1016/j.biopsych.2019.07.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 07/05/2019] [Accepted: 07/15/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Hyperactivation of innate immunity has been implicated in the etiology of mood disorders, including major depressive disorder (MDD). Mesenchymal stromal cells (MSCs) have demonstrated potent immunomodulatory capabilities in the context of chronic inflammatory disease and injury but have yet to be evaluated in stress-based preclinical models of MDD. We sought to test the ability of intravenous MSCs to modulate innate immune activation and behavioral patterns associated with repeated social defeat (RSD). METHODS Murine RSD-induced innate immune activation as well as depressive and anxiety-like behaviors were assessed in unstressed, RSD, and RSD + human MSC groups. Biodistribution and fate studies were performed to inform potential mechanisms of action. RESULTS MSCs reduced stress-induced circulating proinflammatory cytokines, monocytes, neuroinflammation, and depressive and anxiety-like behaviors. Biodistribution analyses indicated that infused MSCs distributed within peripheral organs without homing to the brain. Murine neutrophils targeted MSCs in the lungs within hours of administration. MSCs and recipient neutrophils were cleared by recipient macrophages promoting a switch toward a regulatory phenotype and systemic resolution of inflammation. CONCLUSIONS Peripheral delivery of MSCs modulates central nervous system inflammatory processes and aberrant behavioral patterns in a stress-based rodent model of MDD and anxiety. Recent studies suggest that host immune cell-mediated phagocytosis of MSCs in vivo can trigger an immunomodulatory cascade, resulting in resolution of inflammation. Our data suggest that similar mechanisms may protect distal organs, including the brain, from systemic, stress-induced proinflammatory spikes and may uncover unexpected targets in the periphery for novel or adjunct treatment for a subset of patients with MDD.
Collapse
|
235
|
Shelton KA, Nehete BP, Chitta S, Williams LE, Schapiro SJ, Simmons J, Abee CR, Nehete PN. Effects of Transportation and Relocation on Immunologic Measures in Cynomolgus Macaques ( Macaca fascicularis). JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2019; 58:774-782. [PMID: 31604484 PMCID: PMC6926399 DOI: 10.30802/aalas-jaalas-19-000007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/19/2019] [Accepted: 03/14/2019] [Indexed: 01/08/2023]
Abstract
NHP are a small, but critical, portion of the animals studied in research laboratories. Many NHP are imported or raised at one facility and subsequently moved to another facility for research purposes. To improve our understanding of the effects of transportation and relocation on the NHP immune system, to minimize potential confounds associated with relocation, and to maximize study validity, we examined the phenotype and function of PBMC in cynomolgus macaques (Macaca fascicularis) that were transported approximately 200 miles by road from one facility to another. We evaluated the phenotype of lymphocyte subsets through flow cytometry, mitogen-specific immune responses of PBMC in vitro, and plasma levels of circulating cytokines before transportation, at approximately 24 h after arrival (day 2), and after 30 d of acclimation. Analyses of blood samples revealed that the CD3+ and CD4+ T-cell counts increased significantly, whereas NK+, NKT, and CD14+ CD16+ nonclassical monocyte subsets were decreased significantly on day 2 after relocation compared with baseline. We also noted significantly increased immune cell function as indicated by mitogen-specific proliferative responses and by IFNγ levels on day 2 compared with baseline. After 30 d of acclimation, peripheral blood CD4+ T-cells and monocyte counts were higher than baseline, whereas B-cell numbers were lower. The mitogen-induced responses to LPS and IFNγ production after stimulation with pokeweed mitogen or phytohemagglutinin remained significantly different from baseline. In conclusion, the effects of transportation and relocation on immune parameters in cynomolgus monkeys are significant and do not fully return to baseline values even after 30 d of acclimation.
Collapse
Affiliation(s)
- Kathryn A Shelton
- Department of Comparative Medicine, MD Anderson Cancer Center, University of Texas, Bastrop, Texas
| | - Bharti P Nehete
- Department of Comparative Medicine, MD Anderson Cancer Center, University of Texas, Bastrop, Texas;,
| | - Sriram Chitta
- Department of Comparative Medicine, MD Anderson Cancer Center, University of Texas, Bastrop, Texas
| | - Lawrence E Williams
- Department of Comparative Medicine, MD Anderson Cancer Center, University of Texas, Bastrop, Texas; Graduate School of Biomedical Sciences, University of Texas, Houston, Texas
| | - Steven J Schapiro
- Department of Comparative Medicine, MD Anderson Cancer Center, University of Texas, Bastrop, Texas; Department of Experimental Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Joe Simmons
- Department of Comparative Medicine, MD Anderson Cancer Center, University of Texas, Bastrop, Texas
| | - Christian R Abee
- Department of Comparative Medicine, MD Anderson Cancer Center, University of Texas, Bastrop, Texas
| | - Pramod N Nehete
- Department of Comparative Medicine, MD Anderson Cancer Center, University of Texas, Bastrop, Texas; Graduate School of Biomedical Sciences, University of Texas, Houston, Texas
| |
Collapse
|
236
|
Holmes L, Chinaka C, Elmi H, Deepika K, Pelaez L, Enwere M, Akinola OT, Dabney KW. Implication of Spiritual Network Support System in Epigenomic Modulation and Health Trajectory. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E4123. [PMID: 31717711 PMCID: PMC6862316 DOI: 10.3390/ijerph16214123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 12/16/2022]
Abstract
With challenges in understanding the multifactorial etiologies of disease and individual treatment effect heterogeneities over the past four decades, much has been acquired on how physical, chemical and social environments affect human health, predisposing certain subpopulations to adverse health outcomes, especially the socio-environmentally disadvantaged (SED). Current translational data on gene and adverse environment interaction have revealed how adverse gene-environment interaction, termed aberrant epigenomic modulation, translates into impaired gene expression via messenger ribonucleic acid (mRNA) dysregulation, reflecting abnormal protein synthesis and hence dysfunctional cellular differentiation and maturation. The environmental influence on gene expression observed in most literature includes physical, chemical, physicochemical and recently social environment. However, data are limited on spiritual or religious environment network support systems, which reflect human psychosocial conditions and gene interaction. With this limited information, we aimed to examine the available data on spiritual activities characterized by prayers and meditation for a possible explanation of the nexus between the spiritual network support system (SNSS) as a component of psychosocial conditions, implicated in social signal transduction, and the gene expression correlate. With the intent to incorporate SNSS in human psychosocial conditions, we assessed the available data on bereavement, loss of spouse, loneliness, social isolation, low socio-economic status (SES), chronic stress, low social status, social adversity (SA) and early life stress (ELS), as surrogates for spiritual support network connectome. Adverse human psychosocial conditions have the tendency for impaired gene expression through an up-regulated conserved transcriptional response to adversity (CTRA) gene expression via social signal transduction, involving the sympathetic nervous system (SNS), beta-adrenergic receptors, the hypothalamus-pituitary-adrenal (HPA) axis and the glucocorticoid response. This review specifically explored CTRA gene expression and the nuclear receptor subfamily 3 group C member 1 (NR3C1) gene, a glucocorticoid receptor gene, in response to stress and the impaired negative feedback, given allostatic overload as a result of prolonged and sustained stress and social isolation as well as the implied social interaction associated with religiosity. While more remains to be investigated on psychosocial and immune cell response and gene expression, current data on human models do implicate appropriate gene expression via the CTRA and NR3C1 gene in the SNSS as observed in meditation, yoga and thai-chi, implicated in malignant neoplasm remission. However, prospective epigenomic studies in this context are required in the disease causal pathway, prognosis and survival, as well as cautious optimism in the application of these findings in clinical and public health settings, due to unmeasured and potential confoundings implicated in these correlations.
Collapse
Affiliation(s)
- Laurens Holmes
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Biological Sciences Department, University of Delaware, Newark, DE 19716, USA
- College of Population Health, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Chinacherem Chinaka
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Public Health Department, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Community and Environmental Health Department, Old Dominion University, Norfolk, VA 23507, USA
| | - Hikma Elmi
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
| | - Kerti Deepika
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
| | - Lavisha Pelaez
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
| | - Michael Enwere
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Public Health Department, Walden University, Minneapolis, MN 55401, USA
| | - Olumuyiwa T. Akinola
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Public Health Department, Eastern Virginia Medical School, Norfolk, VA 23507, USA
- Community and Environmental Health Department, Old Dominion University, Norfolk, VA 23507, USA
| | - Kirk W. Dabney
- Nemours Children’s Healthcare System, Nemours Office of Health Equity and Inclusion, Wilmington, DE 19803, USA; (C.C.); (H.E.); (K.D.); (L.P.); (M.E.); (O.T.A.); (K.W.D.)
- Sidney Kimmel Medical School, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
237
|
Social history and exposure to pathogen signals modulate social status effects on gene regulation in rhesus macaques. Proc Natl Acad Sci U S A 2019; 117:23317-23322. [PMID: 31611381 DOI: 10.1073/pnas.1820846116] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Social experience is an important predictor of disease susceptibility and survival in humans and other social mammals. Chronic social stress is thought to generate a proinflammatory state characterized by elevated antibacterial defenses and reduced investment in antiviral defense. Here we manipulated long-term social status in female rhesus macaques to show that social subordination alters the gene expression response to ex vivo bacterial and viral challenge. As predicted by current models, bacterial lipopolysaccharide polarizes the immune response such that low status corresponds to higher expression of genes in NF-κB-dependent proinflammatory pathways and lower expression of genes involved in the antiviral response and type I IFN signaling. Counter to predictions, however, low status drives more exaggerated expression of both NF-κB- and IFN-associated genes after cells are exposed to the viral mimic Gardiquimod. Status-driven gene expression patterns are linked not only to social status at the time of sampling, but also to social history (i.e., past social status), especially in unstimulated cells. However, for a subset of genes, we observed interaction effects in which females who fell in rank were more strongly affected by current social status than those who climbed the social hierarchy. Taken together, our results indicate that the effects of social status on immune cell gene expression depend on pathogen exposure, pathogen type, and social history-in support of social experience-mediated biological embedding in adulthood, even in the conventionally memory-less innate immune system.
Collapse
|
238
|
Sex-specific roles of cellular inflammation and cardiometabolism in obesity-associated depressive symptomatology. Int J Obes (Lond) 2019; 43:2045-2056. [PMID: 31089263 PMCID: PMC6774832 DOI: 10.1038/s41366-019-0375-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 02/06/2019] [Accepted: 03/29/2019] [Indexed: 01/27/2023]
Abstract
BACKGROUND Obesity and depression are complex conditions with stronger comorbid relationships among women than men. Inflammation and cardiometabolic dysfunction are likely mechanistic candidates for increased depression risk, and their prevalence differs by sex. Whether these relationships extend to depressive symptoms is poorly understood. Therefore, we analyzed sex in associations between inflammation and metabolic syndrome (MetS) criteria on depressive symptomatology. Specifically, we examined whether sex positively moderates the relationship between depressive symptoms and inflammation among women, and whether MetS has parallel effects among men. METHODS Depressive symptoms, MetS, and inflammation were assessed in 129 otherwise healthy adults. Depressive symptoms were assessed using Beck Depression Inventory (BDI-Ia). Monocyte inflammation regulation (BARIC) was quantified using flow cytometry measurement of TNF-α suppression by β-agonist. Moderation effects of sex on associations between BARIC, MetS criteria, and BDI were estimated using two-way ANOVA and linear regression, adjusting for BMI, and by sex subgroup analyses. RESULTS Obese individuals reported more depressive symptoms. Sex did not formally moderate this relationship, though BDI scores tended to differ by BMI among women, but not men, in subgroup analysis. Poorer inflammation control and higher MetS criteria were correlated with somatic depressive symptoms. Sex moderated associations between MetS criteria and somatic symptoms; among men, MetS criteria predicted somatic symptoms, not among women. Subgroup analysis further indicated that poorer inflammation control tended to be associated with higher somatic symptoms in women. CONCLUSIONS These results indicate that obesity-related inflammation and MetS factors have sex-specific effects on depressive symptoms in a non-clinical population. Although pathophysiological mechanisms underlying sex differences remain to be elucidated, our findings suggest that distinct vulnerabilities to depressive symptoms exist between women and men, and highlight the need to consider sex as a key biological variable in obesity-depression relationships. Future clinical studies on comorbid obesity and depression should account for sex, which may optimize therapeutic strategies.
Collapse
|
239
|
van de Wouw M, Boehme M, Dinan TG, Cryan JF. Monocyte mobilisation, microbiota & mental illness. Brain Behav Immun 2019; 81:74-91. [PMID: 31330299 DOI: 10.1016/j.bbi.2019.07.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/12/2019] [Accepted: 07/18/2019] [Indexed: 12/13/2022] Open
Abstract
The gastrointestinal microbiome has emerged as a key player in regulating brain and behaviour. This has led to the strategy of targeting the gut microbiota to ameliorate disorders of the central nervous system. Understanding the underlying signalling pathways in which the microbiota impacts these disorders is crucial for the development of future therapeutics for improving CNS functionality. One of the major pathways through which the microbiota influences the brain is the immune system, where there is an increasing appreciation for the role of monocyte trafficking in regulating brain homeostasis. In this review, we will shed light on the role of monocyte trafficking as a relay of microbiota signals in conditions where the central nervous system is in disorder, such as stress, peripheral inflammation, ageing, traumatic brain injury, stroke, multiple sclerosis, Alzheimer's disease and Parkinson's disease. We also cover how the gastrointestinal microbiota is implicated in these mental illnesses. In addition, we aim to discuss how the monocyte system can be modulated by the gut microbiota to mitigate disorders of the central nervous system, which will lead to novel microbiota-targeted strategies.
Collapse
Affiliation(s)
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
240
|
Yuan N, Chen Y, Xia Y, Dai J, Liu C. Inflammation-related biomarkers in major psychiatric disorders: a cross-disorder assessment of reproducibility and specificity in 43 meta-analyses. Transl Psychiatry 2019; 9:233. [PMID: 31534116 PMCID: PMC6751188 DOI: 10.1038/s41398-019-0570-y] [Citation(s) in RCA: 251] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 05/24/2019] [Indexed: 12/15/2022] Open
Abstract
Inflammation is a natural defence response of the immune system against environmental insult, stress and injury, but hyper- and hypo-inflammatory responses can trigger diseases. Accumulating evidence suggests that inflammation is involved in multiple psychiatric disorders. Using inflammation-related factors as biomarkers of psychiatric disorders requires the proof of reproducibility and specificity of the changes in different disorders, which remains to be established. We performed a cross-disorder study by systematically evaluating the meta-analysis results of inflammation-related factors in eight major psychiatric disorders, including schizophrenia (SCZ), bipolar disorder (BD), autism spectrum disorder (ASD), major depression disorder (MDD), post-trauma stress disorder (PTSD), sleeping disorder (SD), obsessive-compulsive disorder (OCD) and suicide. A total of 43 meta-analyses involving 704 publications on 44 inflammation-related factors were included in the study. We calculated the effect size and statistical power for every inflammation-related factor in each disorder. Our analyses showed that well-powered case-control studies provided more consistent results than underpowered studies when one factor was meta-analysed by different researchers. After removing underpowered studies, 30 of the 44 inflammation-related factors showed significant alterations in at least one disorder based on well-powered meta-analyses. Eleven of them changed in patients of more than two disorders when compared with the controls. A few inflammation-related factors showed unique changes in specific disorders (e.g., IL-4 increased in BD, decreased in suicide, but had no change in MDD, ASD, PTSD and SCZ). MDD had the largest number of changes while SD has the least. Clustering analysis showed that closely related disorders share similar patterns of inflammatory changes, as genome-wide genetic studies have found. According to the effect size obtained from the meta-analyses, 13 inflammation-related factors would need <50 cases and 50 controls to achieve 80% power to show significant differences (p < 0.0016) between patients and controls. Changes in different states of MDD, SCZ or BD were also observed in various comparisons. Studies comparing first-episode SCZ to controls may have more reproducible findings than those comparing pre- and post-treatment results. Longitudinal, system-wide studies of inflammation regulation that can differentiate trait- and state-specific changes will be needed to establish valuable biomarkers.
Collapse
Affiliation(s)
- Ning Yuan
- Department of Psychiatry, The Second Xiangya Hospital; Mental health Institute of the Second Xiangya Hospital; National Clinical Research Center on Mental Disorders; National Technology Institute on Mental Disorders, Central South University, Changsha, Hunan, China
- Department of Psychiatry, Hunan Provincial Brain Hospital; Clinical Research Center for Mental Behavioral Disorder in Hunan Province, Clinical Medical School of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yu Chen
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yan Xia
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jiacheng Dai
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Chunyu Liu
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China.
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA.
- School of Psychology, Shaanxi Normal University, Xi'an, Shaanxi, China.
| |
Collapse
|
241
|
Pfau ML, Menard C, Cathomas F, Desland F, Kana V, Chan KL, Shimo Y, LeClair K, Flanigan ME, Aleyasin H, Walker DM, Bouchard S, Mack M, Hodes GE, Merad MM, Russo SJ. Role of Monocyte-Derived MicroRNA106b∼25 in Resilience to Social Stress. Biol Psychiatry 2019; 86:474-482. [PMID: 31101319 PMCID: PMC6717005 DOI: 10.1016/j.biopsych.2019.02.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 02/13/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Clinical studies suggest that heightened peripheral inflammation contributes to the pathogenesis of stress-related disorders, including major depressive disorder. However, the molecular mechanisms within peripheral immune cells that mediate enhanced stress vulnerability are not well known. Because microRNAs (miRs) are important regulators of immune response, we sought to examine their role in mediating inflammatory and behavioral responses to repeated social defeat stress (RSDS), a mouse model of stress vulnerability that produces susceptible and resilient phenotypes. METHODS We isolated Ly6chigh monocytes via fluorescence-activated cell sorting in the blood of susceptible and resilient mice following RSDS and profiled miR expression via quantitative real-time polymerase chain reaction. Bone marrow chimeric mice were generated to confirm a causal role of the miR-106b∼25 cluster in bone marrow-derived leukocytes in mediating stress resilience versus susceptibility. RESULTS We found that RSDS produces an increase in circulating Ly6chigh inflammatory monocytes in both susceptible and resilient mice. We next investigated whether intrinsic leukocyte posttranscriptional mechanisms contribute to individual differences in stress response and the resilient phenotype. Of the miRs profiled in our panel, eight were significantly regulated by RSDS within Ly6chigh monocytes, including miR-25-3p, a member of the miR-106b∼25 cluster. Selective knockout of the miR-106b∼25 cluster in peripheral leukocytes promoted behavioral resilience to RSDS. CONCLUSIONS Our results identify the miR-106b∼25 cluster as a key regulator of stress-induced inflammation and depression that may represent a novel therapeutic target for drug development.
Collapse
Affiliation(s)
- Madeline L Pfau
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Caroline Menard
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry and Neuroscience, Faculty of Medicine and Cervo Brain Research Center, Université Laval, Quebec City, Quebec, Canada
| | - Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fiona Desland
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Oncological Science, Tisch Cancer Institute and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Veronika Kana
- Department of Oncological Science, Tisch Cancer Institute and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kenny L Chan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yusuke Shimo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Katherine LeClair
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Meghan E Flanigan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Hossein Aleyasin
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Deena M Walker
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sylvain Bouchard
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Matthias Mack
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Georgia E Hodes
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Miriam M Merad
- Department of Oncological Science, Tisch Cancer Institute and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
242
|
Cathomas F, Murrough JW, Nestler EJ, Han MH, Russo SJ. Neurobiology of Resilience: Interface Between Mind and Body. Biol Psychiatry 2019; 86:410-420. [PMID: 31178098 PMCID: PMC6717018 DOI: 10.1016/j.biopsych.2019.04.011] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/26/2019] [Accepted: 04/05/2019] [Indexed: 12/12/2022]
Abstract
Stress-related neuropsychiatric disorders, such as major depressive disorder and posttraumatic stress disorder, exact enormous socioeconomic and individual consequences. Resilience, the process of adaptation in the face of adversity, is an important concept that is enabling the field to understand individual differences in stress responses, with the hope of harnessing this information for the development of novel therapeutics that mimic the body's natural resilience mechanisms. This review provides an update on the current state of research of the neurobiological mechanisms of stress resilience. We focus on physiological and transcriptional adaptations of specific brain circuits, the role of cellular and humoral factors of the immune system, the gut microbiota, and changes at the interface between the brain and the periphery, the blood-brain barrier. We propose viewing resilience as a process that requires the integration of multiple central and peripheral systems and that elucidating the underlying neurobiological mechanisms will ultimately lead to novel therapeutic options.
Collapse
Affiliation(s)
- Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - James W Murrough
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ming-Hu Han
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
243
|
Knight JM, Rizzo JD, Wang T, He N, Logan BR, Spellman SR, Lee SJ, Verneris MR, Arevalo JMG, Cole SW. Molecular Correlates of Socioeconomic Status and Clinical Outcomes Following Hematopoietic Cell Transplantation for Leukemia. JNCI Cancer Spectr 2019; 3:pkz073. [PMID: 31763620 PMCID: PMC6859844 DOI: 10.1093/jncics/pkz073] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/24/2019] [Accepted: 09/09/2019] [Indexed: 01/08/2023] Open
Abstract
Background Clinical outcomes among allogeneic hematopoietic cell transplant (HCT) recipients are negatively affected by low socioeconomic status (SES), yet the biological mechanisms accounting for this health disparity remain to be elucidated. Among unrelated donor HCT recipients with acute myelogenous leukemia, one recent pilot study linked low SES to increased expression of a stress-related gene expression profile known as the conserved transcriptional response to adversity (CTRA) in peripheral blood mononuclear cells, which involves up-regulation of pro-inflammatory genes and down-regulation of genes involved in type I interferon response and antibody synthesis. Methods This study examined these relationships using additional measures in a larger archival sample of 261 adults who received an unrelated donor HCT for acute myelogenous leukemia to 1) identify cellular and molecular mechanisms involved in SES-related differences in pre-transplant leukocyte transcriptome profiles, and 2) evaluate pre-transplant CTRA biology associations with clinical outcomes through multivariable analysis controlling for demographic-, disease-, and transplant-related covariates. Results Low SES individuals showed increases in classic monocyte activation and pro-inflammatory transcription control pathways as well as decreases in activation of nonclassic monocytes, all consistent with the CTRA biological pattern. Transplant recipients in the highest or lowest quartiles of the CTRA pro-inflammatory gene component had a more than 2-fold elevated hazard of relapse (hazard ratio [HR] = 2.47, 95% confidence interval [CI] = 1.44 to 4.24), P = .001; HR = 2.52, 95% CI = 1.46 to 4.34, P = .001) and more than 20% reduction in leukemia-free survival (HR = 1.57, 95% CI = 1.08 to 2.28, P = .012; HR = 1.49, 95% CI = 1.04 to 2.15, P = .03) compared with the middle quartiles. Conclusions These findings identify SES- and CTRA-associated myeloid- and inflammation-related transcriptome signatures in recipient pre-transplant blood samples as a potential novel predictive biomarker of HCT-related clinical outcomes.
Collapse
Affiliation(s)
| | - J Douglas Rizzo
- See the Notes section for the full list of authors' affiliations
| | - Tao Wang
- See the Notes section for the full list of authors' affiliations
| | - Naya He
- See the Notes section for the full list of authors' affiliations
| | - Brent R Logan
- See the Notes section for the full list of authors' affiliations
| | | | - Stephanie J Lee
- See the Notes section for the full list of authors' affiliations
| | | | | | - Steve W Cole
- See the Notes section for the full list of authors' affiliations
| |
Collapse
|
244
|
Bower JE, Kuhlman KR, Haydon MD, Boyle CC, Radin A. Cultivating a healthy neuro‐immune network: A health psychology approach. SOCIAL AND PERSONALITY PSYCHOLOGY COMPASS 2019; 13. [PMID: 37008404 PMCID: PMC10062207 DOI: 10.1111/spc3.12498] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The field of psychoneuroimmunology (PNI) examines interactions among psychological and behavioral states, the brain, and the immune system. Research in PNI has elegantly documented effects of stress at multiple levels of the neuro-immune network, with profound implications for both physical and mental health. In this review, we consider how the neuro-immune network might be influenced by "positive" psychological and behavioral states, focusing on positive affect, eudaimonic well-being, physical activity, and sleep. There is compelling evidence that these positive states and behaviors are associated with changes in immune activity in the body, including reductions in peripheral inflammatory processes relevant for physical health. Growing evidence from animal models also suggests effects of positive states on immune cells in the brain and the blood-brain barrier, which then impact critical aspects of mood, cognition, and behavior. Tremendous advances are being made in our understanding of neuro-immune dynamics; one of the central goals of this review is to highlight recent preclinical research in this area and consider how we can leverage these findings to investigate and cultivate a healthy neuro-immune network in humans.
Collapse
Affiliation(s)
| | - Kate R. Kuhlman
- University of California Los Angeles
- University of California Irvine
| | | | | | | |
Collapse
|
245
|
Abstract
Cardiac ageing manifests as a decline in function leading to heart failure. At the cellular level, ageing entails decreased replicative capacity and dysregulation of cellular processes in myocardial and nonmyocyte cells. Various extrinsic parameters, such as lifestyle and environment, integrate important signalling pathways, such as those involving inflammation and oxidative stress, with intrinsic molecular mechanisms underlying resistance versus progression to cellular senescence. Mitigation of cardiac functional decline in an ageing organism requires the activation of enhanced maintenance and reparative capacity, thereby overcoming inherent endogenous limitations to retaining a youthful phenotype. Deciphering the molecular mechanisms underlying dysregulation of cellular function and renewal reveals potential interventional targets to attenuate degenerative processes at the cellular and systemic levels to improve quality of life for our ageing population. In this Review, we discuss the roles of extrinsic and intrinsic factors in cardiac ageing. Animal models of cardiac ageing are summarized, followed by an overview of the current and possible future treatments to mitigate the deleterious effects of cardiac ageing.
Collapse
|
246
|
Gillespie SL, Cole SW, Christian LM. Early adversity and the regulation of gene expression: Implications for prenatal health. Curr Opin Behav Sci 2019; 28:111-118. [PMID: 31815157 PMCID: PMC6897329 DOI: 10.1016/j.cobeha.2019.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Early life, including prenatal development and childhood, is a period of sensitivity, with potential for developmental programming under conditions of adversity. The intergenerational effects of early adversity have received attention, most often studied in relation to fetal development according to maternal exposures. Less often considered but critically important is the effect of early adversity on future prenatal risk (e.g., risk for preeclampsia, preterm birth), which threatens the health of mother and infant. The body's ability to turn collections of genes "on" or "off" across a range of tissues via receptor-driven transcription factors and epigenetic mechanisms (i.e., chemical modifications to the genome) in response to the perceived environment may help to explain such associations. This review aims to summarize discoveries surrounding the effects of early adversity on gene expression, emphasizing prenatal populations. First, we review findings from gene expression studies examining the effects of early adversity on various tissues known to contribute to prenatal health in adulthood. Next, we review several gene regulatory mechanisms thought to underlie differences in gene expression. Finally, we discuss potential implications for prenatal risk among early adversity-exposed mothers according to our current understanding of the biology that contributes to the development of prenatal syndromes.
Collapse
Affiliation(s)
| | - Steve W Cole
- Department of Psychiatry & Biobehavioral Sciences and Medicine, UCLA School of Medicine, Los Angeles, CA
| | - Lisa M Christian
- Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH
- The Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH
| |
Collapse
|
247
|
Thames AD, Irwin MR, Breen EC, Cole SW. Experienced discrimination and racial differences in leukocyte gene expression. Psychoneuroendocrinology 2019; 106:277-283. [PMID: 31029930 PMCID: PMC6589103 DOI: 10.1016/j.psyneuen.2019.04.016] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 02/07/2019] [Accepted: 04/18/2019] [Indexed: 01/03/2023]
Abstract
Racial disparities in health outcomes between African Americans and European Americans have been well-documented, but not fully understood. Chronic inflammation contributes to several of the diseases showing racial disparities (e.g., Human Immunodeficiency Virus [HIV]), and racial differences in stress exposure (e.g., experiences of racial discrimination) that stimulate pro-inflammatory processes that may contribute to differential health outcomes. We performed a cross-sectional bioinformatic analyses relating perceived discrimination (as measured by the Perceived Ethnic Discrimination Questionnaire [PED-Q]) to the activity of pro-inflammatory, neuroendocrine, and antiviral transcription control pathways relevant to the conserved transcriptional response to adversity (CTRA) in peripheral blood leukocytes. Subjects were 71 individuals (37 HIV-seropositive (HIV+); 34 HIV-seronegative (HIV-)) (mean age = 53 years, range 27-63), who self-identified either as African American/Black (n = 48) or European American/White (n = 23). This provided the opportunity to examine the independent effects of race and HIV, as well as the modifying role of perceived discrimination on pathways involved in CTRA. Exploratory analysis examined the interactive effects of HIV and race on pathways involved in CTRA. Relative to European Americans, African Americans showed increased activity of two key pro-inflammatory transcription control pathways (NF- кB and AP-1) and two stress-responsive signaling pathways (CREB and glucocorticoid receptor); these effects did not differ significantly as a function of HIV infection (HIV x Race interaction, all p > .10). Results suggested that differences in experiences of racial discrimination could potentially account for more than 50% of the total race-related difference in pro-inflammatory transcription factor activity. In sum, differential exposure to racial discrimination may contribute to racial disparities in health outcomes in part by activating threat-related molecular programs that stimulate inflammation and contribute to increased risk of chronic illnesses.
Collapse
Affiliation(s)
- April D Thames
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, United States.
| | - Michael R Irwin
- Cousins Center for Psychoneuroimmunology, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, United States
| | - Elizabeth C Breen
- Cousins Center for Psychoneuroimmunology, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, United States
| | - Steve W Cole
- Cousins Center for Psychoneuroimmunology, Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, United States
| |
Collapse
|
248
|
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
|
249
|
Novel Treatment Targets Based on Insights in the Etiology of Depression: Role of IL-6 Trans-Signaling and Stress-Induced Elevation of Glutamate and ATP. Pharmaceuticals (Basel) 2019; 12:ph12030113. [PMID: 31362361 PMCID: PMC6789839 DOI: 10.3390/ph12030113] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/10/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022] Open
Abstract
Inflammation and psychological stress are risk factors for major depression and suicide. Both increase central glutamate levels and activate the hypothalamic-pituitary-adrenal axis and the sympathetic nervous system. Both factors also affect the function of the chloride transporters, Na-K-Cl-cotransporter-1 (NKCC1) and K-Cl-cotransporter-2 (KCC2), and provoke interleukin-6 (IL-6) trans-signaling. This leads to measurable increases in circulating corticosteroids, catecholamines, anxiety, somatic and psychological symptoms, and a decline in cognitive functions. Recognition of the sequence of pathological events allows the prediction of novel targets for therapeutic intervention. Amongst others, these include blockade of the big-K potassium channel, blockade of the P2X4 channel, TYK2-kinase inhibition, noradrenaline α2B-receptor antagonism, nicotinic α7-receptor stimulation, and the Sgp130Fc antibody. A better understanding of downstream processes evoked by inflammation and stress also allows suggestions for tentatively better biomarkers (e.g., SERPINA3N, MARCKS, or 13C-tryptophan metabolism).
Collapse
|
250
|
Leite F, Ribeiro L. Dopaminergic Pathways in Obesity-Associated Inflammation. J Neuroimmune Pharmacol 2019; 15:93-113. [PMID: 31317376 DOI: 10.1007/s11481-019-09863-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 07/02/2019] [Indexed: 12/11/2022]
|