Hostname: page-component-848d4c4894-5nwft Total loading time: 0 Render date: 2024-05-02T15:37:16.279Z Has data issue: false hasContentIssue false

Challenges in researching the immune pathways between early life adversity and psychopathology

Published online by Cambridge University Press:  11 January 2021

Brie Reid*
Affiliation:
Institute of Child Development, University of Minnesota, Minneapolis, USA
Andrea Danese
Affiliation:
Institute of Child Development, University of Minnesota, Minneapolis, USA Social, Genetic and Developmental Psychiatry Centre, King's College London, London, UK Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK National and Specialist CAMHS Clinic for Trauma, Anxiety, and Depression, South London and Maudsley NHS Foundation Trust, London, UK
*
Author for Correspondence: Brie Reid, Institute of Child Development, University of Minnesota; Minneapolis, MN. Institute of Child Development, 51 East River Road, Minneapolis, MN55455; E-mail: reidx189@umn.edu.
Rights & Permissions [Opens in a new window]

Abstract

Exposure to childhood adversity is a critical risk factor for the development of psychopathology. A growing field of research examines how exposure to childhood adversity is translated into biological risk for psychopathology through alterations in immune system functioning, most notably heightened levels of inflammation biomarkers. Though our knowledge about how childhood adversity can instantiate biological risk for psychopathology is growing, there remain many challenges and gaps in the field to understand how inflammation from childhood adversity contributes to psychopathology. This paper reviews research on the inflammatory outcomes arising from childhood adversity and presents four major challenges that future research must address: (a) the measurement of childhood adversity, (b) the measurement of inflammation, (c) the identification of mediators between childhood adversity and inflammation, and (d) the identification of moderators of inflammatory outcomes following childhood adversity. We discuss synergies and inconsistencies in the literature to summarize the current understanding of the association between childhood adversity, a proinflammatory phenotype, and the biological risk for psychopathology. We discuss the clinical implications of the inflammatory links between childhood adversity and psychopathology, including possibilities for intervention. Finally, this review conclude by delineates future directions for research, including issues of how best to detect, prevent, and understand these “hidden wounds” of childhood adversity.

Type
Special Section 1: 2019 Minnesota Symposium on Child Psychology
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
Copyright © The Author(s), 2020. Published by Cambridge University Press

Introduction

Childhood adversity is widespread and associated with increased risk for mental health problems and psychopathology in adulthood (Danese, Reference Danese2020; Humphreys & Zeanah, Reference Humphreys and Zeanah2015; McLaughlin, Reference McLaughlin2016; Teicher & Samson, Reference Teicher and Samson2013). Childhood adversities are defined as exposures to circumstances that represent a deviation from the expectable environment, either chronic or severe and acute, during childhood or adolescence that require significant adaptation by an average child (McLaughlin, Sheridan, & Lambert, Reference McLaughlin, Sheridan and Lambert2014). We will center our discussion of early life adversity (ELA) around the construct of child victimization, which includes experiences of maltreatment by an adult (i.e., emotional, physical, or sexual abuse, and physical or emotional neglect) and bullying by peers (Danese & Lewis, Reference Danese and Lewis2017). ELA is associated with greater risk for developing mental disorders early and across the life course in humans, and the odds of developing psychopathology increase as exposure to adversity increases (Kessler, Davis, & Kendler, Reference Kessler, Davis and Kendler1997; Khandaker, Pearson, Zammit, Lewis, & Jones, Reference Khandaker, Pearson, Zammit, Lewis and Jones2010; Schaefer et al., Reference Schaefer, Moffitt, Arseneault, Danese, Fisher, Houts and Caspi2018). Animal models of ELA experimentally show that the associations between early contexts of stress and later emotional and behavioral abnormalities is likely causal (Cryan & Holmes, Reference Cryan and Holmes2005; Francis, Diorio, Liu, & Meaney, Reference Francis, Diorio, Liu and Meaney1999; Harlow, Dodsworth, & Harlow, Reference Harlow, Dodsworth and Harlow1965; Hinde & Spencer-Booth, Reference Hinde and Spencer-Booth1971; Levine, Chevalier, & Korchin, Reference Levine, Chevalier and Korchin1956; Sanchez, Ladd, & Plotsky, Reference Sanchez, Ladd and Plotsky2001; Suomi, Reference Suomi1997).

While exposure to ELA increases the odds of many psychiatric disorders, the association between ELA and different types of psychopathology is generally nonspecific (Green et al., Reference Green, McLaughlin, Berglund, Gruber, Sampson, Zaslavsky and Kessler2010; Kessler et al., Reference Kessler, Davis and Kendler1997; Kessler et al., Reference Kessler, McLaughlin, Green, Gruber, Sampson, Zaslavsky and Williams2010; McLaughlin et al., Reference McLaughlin, Greif Green, Gruber, Sampson, Zaslavsky and Kessler2012; Schaefer et al., Reference Schaefer, Moffitt, Arseneault, Danese, Fisher, Houts and Caspi2018). For example, childhood maltreatment predicts several psychiatric disorders, including depression, bipolar disorder, posttraumatic stress disorder (PTSD), and schizophrenia (Gilbert et al., Reference Gilbert, Widom, Browne, Fergusson, Webb and Janson2009; Varese et al., Reference Varese, Smeets, Drukker, Lieverse, Lataster, Viechtbauer and Bentall2012; Widom, Reference Widom1999). Childhood maltreatment is associated with an unfavorable course of mental illness, higher rates of comorbid disorders, and more complex presentation of symptoms (Agnew-Blais & Danese, Reference Agnew-Blais and Danese2016; Maercker et al., Reference Maercker, Brewin, Bryant, Cloitre, van Ommeren, Jones and Reed2013; Nanni, Uher, & Danese, Reference Nanni, Uher and Danese2012). In addition, ELA is associated with many physical health outcomes, including metabolic syndrome, cardiovascular disease, neurodegenerative diseases, cancer, asthma, and aging (Scrivo, Vasile, Bartosiewicz, & Valesini, Reference Scrivo, Vasile, Bartosiewicz and Valesini2011). Given the nonspecific associations and the co-occurring risk for psychopathology and physical health disorders, inflammation has been proposed as the “common soil” by which adversity becomes biologically embedded to influence later psychological and physical functioning (Scrivo et al., Reference Scrivo, Vasile, Bartosiewicz and Valesini2011). The immune system, via inflammation, is increasingly recognized as a pathway to developing psychopathology (Danese & Baldwin, Reference Danese and Baldwin2017; Nusslock & Miller, Reference Nusslock and Miller2016). In longitudinal studies of depression, high levels of inflammation predict an increased risk of developing depression, poor response to treatment (Strawbridge et al., Reference Strawbridge, Arnone, Danese, Papadopoulos, Herane Vives and Cleare2015), and an unfavorable course of illness (Ford & Erlinger, Reference Ford and Erlinger2004). In bipolar disorder, high levels of inflammation predict poor treatment response (Mondelli et al., Reference Mondelli, Ciufolini, Belvederi Murri, Bonaccorso, Di Forti, Giordano and Dazzan2015). All of these associations echo the negative psychopathology outcomes in individuals exposed to ELA (Danese & Baldwin, Reference Danese and Baldwin2017).

Research continues to find that ELA confers specific brain, behavioral, and physiological vulnerabilities to inflammation. There are several excellent reviews that outline the links between ELA, inflammation, and psychopathology (Danese & Baldwin, Reference Danese and Baldwin2017; Danese & Lewis, Reference Danese and Lewis2017). This area is an exciting frontier in the understanding of the etiology and incidence of mental illness arising from ELA. Thus, with the availability of multiple reviews, meta-analyses, and new data, this review outlines the outstanding questions on in the links from ELA to inflammation to psychopathology. We provide an overview of the current evidence that connects ELA to increased inflammation and subsequent risk for psychopathology. Then, we identify four challenges for the field going forward. The first is the issue of measuring ELA, which includes the type and timing, dimensional approaches to the construct of ELA, and construct invariance across longitudinal studies. The second challenge is measuring inflammation itself, which includes questions about biomarker selection and tissue specificity in the study of ELA, inflammation, and psychopathology. Third, we review a number of proposed mechanistic pathways that could mediate the relationship between childhood adversity and inflammation to ultimately prevent ELA-exposed children from developing increased inflammation and psychopathology. As a great deal of heterogeneity exists in the literature on ELA and inflammation, we propose that the fourth challenge of the field is a better understanding of moderators at play. We put forward the hypotheses that differences in an individual's age at the inflammation assessment, sex, and population context can explain the heterogeneity in outcomes. We conclude with a discussion of the clinical and intervention implications as the field continues to mature.

Inflammation and psychopathology

The immune system consists of two interconnected branches: innate and adaptive immunity. These branches detect and eliminate molecules and cells that represent a threat to our body, including foreign antigens, altered self-antigens, or cellular damage. Inflammation is part of the innate immune system and works to protect against pathogens and repair tissue in contexts of illness, injury, or infection. Innate immunity serves as the first line of defense and is evolutionally older than adaptive immunity (Medzhitov, Reference Medzhitov2008). The innate immune system is composed of immune cells, such as monocytes, macrophages, and dendritic cells, that constantly circulate in the body and detect a wide variety of pathogens. When activated, these cells begin a rapid (i.e., minutes to hours) cascade of processes to contain infection and promote healing (Medzhitov, Reference Medzhitov2008). Inflammatory cytokines promote vascular permeability and cellular adhesion so that immune cells can leave the blood vessels, migrate to tissues, and eliminate pathogens. Proinflammatory cytokines signal sites of injury or infection to recruit immune cells to the site of inflammatory activity (Murphy, Reference Murphy2011). However, if the regulatory processes in immunity fail, this this can result in a prolonged, chronic, and nonresolving inflammatory response associated with cellular and tissue damage and poor mental and physical health outcomes (Nusslock & Miller, Reference Nusslock and Miller2016). Low-grade inflammation is frequently assessed in human studies using the acute-phase protein C-reactive protein (CRP) and the proinflammatory cytokine interleukin-6 (IL-6), both of which can be measured in blood.

Inflammation is implicated in the pathogenesis of psychopathology in nonhuman animal models and human studies. In experimental animal models, the administration of bacterial cell components (e.g., lipopolysaccharide (LPS)) or proinflammatory cytokines manipulates the organism's inflammatory state and produce symptoms that mirror psychopathology (e.g., disruptions in sleep, food and water intake, social engagement, motor activity, and cognition) (Dantzer, O'Connor, Freund, Johnson, & Kelley, Reference Dantzer, O'Connor, Freund, Johnson and Kelley2008). These symptoms are worse in mice deficient in anti-inflammatory cytokines but improve if mice are subsequently administered anti-inflammatory cytokines (Dantzer et al., Reference Dantzer, O'Connor, Freund, Johnson and Kelley2008). Experimental studies in humans have replicated these findings. Early studies found that the experimental administration of proinflammatory cytokines produced a clinical response that resembles that of major depression (Smith, Reference Smith1991). Later studies demonstrate that the administration of proinflammatory mediators induces depressive-like symptoms in healthy participants and cancer patients (Musselman et al., Reference Musselman, Lawson, Gumnick, Manatunga, Penna, Goodkin and Miller2001; Reichenberg et al., Reference Reichenberg, Yirmiya, Schuld, Kraus, Haack, Morag and Pollmacher2001).

Human studies of depression, bipolar disorder, PTSD, and schizophrenia show evidence of a link between inflammation and psychopathology development (Berk et al., Reference Berk, Kapczinski, Andreazza, Dean, Giorlando, Maes and Malhi2011; Dargel, Godin, Kapczinski, Kupfer, & Leboyer, Reference Dargel, Godin, Kapczinski, Kupfer and Leboyer2015; Leboyer et al., Reference Leboyer, Soreca, Scott, Frye, Henry, Tamouza and Kupfer2012; Miller, Buckley, Seabolt, Mellor, & Kirkpatrick, Reference Miller, Buckley, Seabolt, Mellor and Kirkpatrick2011a; Modabbernia, Taslimi, Brietzke, & Ashrafi, Reference Modabbernia, Taslimi, Brietzke and Ashrafi2013). Of these, the association between inflammation and psychopathology has been best characterized in the case of depression (Miller & Raison, Reference Miller and Raison2016). Patients with depression had immune cell profiles characterized by systemic inflammation (Maes et al., Reference Maes, Lambrechts, Bosmans, Jacobs, Suy, Vandervorst and Raus1992). Moreover, a meta-analysis of cross-sectional studies that investigate the differences in inflammation in depressed adults and healthy controls found that depression is characterized by small elevations of inflammatory biomarkers in the blood (Howren, Lamkin, & Suls, Reference Howren, Lamkin and Suls2009). In addition, anti-inflammatory medications have been found to have antidepressant effects in trial with a subgroup of depressed patients with high baseline levels of inflammation (Raison et al., Reference Raison, Rutherford, Woolwine, Shuo, Schettler, Drake and Miller2013) and in a meta-analysis of trials (Kohler et al., Reference Kohler, Benros, Nordentoft, Farkouh, Iyengar, Mors and Krogh2014). The group differences between inflammation in depressed patients versus controls is likely due to bidirectional associations between depression and inflammation (Danese & Baldwin, Reference Danese and Baldwin2017; Matthews et al., Reference Matthews, Schott, Bromberger, Cyranowski, Everson-Rose and Sowers2010). Concordantly, longitudinal associations between inflammation and subsequent psychopathology are reported in participants with depression (Valkanova, Ebmeier, & Allan, Reference Valkanova, Ebmeier and Allan2013), psychosis (Khandaker et al., Reference Khandaker, Pearson, Zammit, Lewis and Jones2014), and PTSD (Eraly et al., Reference Eraly, Nievergelt, Maihofer, Barkauskas, Biswas and Agorastos2014; Michopoulos et al., Reference Michopoulos, Rothbaum, Jovanovic, Almli, Bradley, Rothbaum and Ressler2015).

Proposed mechanisms of inflammation and psychopathology

While inflammation contributes positively to brain development, high levels can result in changes at multiple levels of brain function and increase risk for psychopathology. In rodents and nonhuman primates, experimental studies show that infection and systemic inflammation in the prenatal and neonatal periods lead to long-term impairments in learning, memory, and attention (Bilbo & Schwarz, Reference Bilbo and Schwarz2009; Knuesel et al., Reference Knuesel, Chicha, Britschgi, Schobel, Bodmer, Hellings and Prinssen2014; Patterson, Reference Patterson2009; Short et al., Reference Short, Lubach, Karasin, Olsen, Styner, Knickmeyer and Coe2010). Observational studies in humans demonstrate links between prenatal infection and later risk for neurodevelopmental disorders, including schizophrenia and autism (Atladottir et al., Reference Atladottir, Thorsen, Ostergaard, Schendel, Lemcke, Abdallah and Parner2010; Deykin & MacMahon, Reference Deykin and MacMahon1979; Khandaker, Zimbron, Lewis, & Jones, Reference Khandaker, Zimbron, Lewis and Jones2013; Mednick, Machon, Huttunen, & Bonett, Reference Mednick, Machon, Huttunen and Bonett1988). Experimental activation of the peripheral system with LPS and other proinflammatory stimulants has significant effects on brain functioning in humans (Schedlowski, Engler, & Grigoleit, Reference Schedlowski, Engler and Grigoleit2014). These effects include changes in cognition with the reduction of verbal and nonverbal memory functions (Reichenberg et al., Reference Reichenberg, Yirmiya, Schuld, Kraus, Haack, Morag and Pollmacher2001) and changes in positive valence systems with the reduction of ventral striatum responses to reward (Capuron et al., Reference Capuron, Pagnoni, Drake, Woolwine, Spivey, Crowe and Miller2012; Eisenberger et al., Reference Eisenberger, Berkman, Inagaki, Rameson, Mashal and Irwin2010). These effects include changes in negative valence systems, with increased amygdala activity in response to socially threatening stimuli (fear faces) (Inagaki, Muscatell, Irwin, Cole, & Eisenberger, Reference Inagaki, Muscatell, Irwin, Cole and Eisenberger2012), reduced connectivity of the subgenual anterior cingulate cortex (sACC) to the amygdala, and increases in the activity within the sACC during emotional face processing (Harrison et al., Reference Harrison, Brydon, Walker, Gray, Steptoe and Critchley2009). Inflammation can affect brain and behavior mechanistically through (a) brain neurochemistry, (b) brain development, (c) microglial priming, and (d) neuroendocrine programming (i.e., hypothalamic–pituitary–adrenal; HPA). We briefly review these mechanistic pathways and direct the reader to Danese and Lewis (Reference Danese and Lewis2017) for a more comprehensive review.

First, inflammation can affect brain neurochemistry through monoamine and glutamate metabolism. Inflammatory cytokines can affect monoamine transmission through reduced monoamine synthesis and increased expression of the presynaptic transporter, reducing monoamine availability in the brain (Miller & Raison, Reference Miller and Raison2015). Inflammatory cytokines can increase glutamine transmission through reduced expression of the presynaptic transporter, thereby increasing glutamate release and availability in the brain (Miller & Raison, Reference Miller and Raison2016).

Second, inflammation plays an integral role in brain development. Prenatally, the immune system affects cell proliferation and migration (Boulanger, Reference Boulanger2009). Postnatally, the immune system affects brain synaptogenesis (Chalasani, Sabelko, Sunshine, Littman, & Raper, Reference Chalasani, Sabelko, Sunshine, Littman and Raper2003; Lieberam, Agalliu, Nagasawa, Ericson, & Jessell, Reference Lieberam, Agalliu, Nagasawa, Ericson and Jessell2005; Tran & Miller, Reference Tran and Miller2003), synaptic refinement (Beattie et al., Reference Beattie, Stellwagen, Morishita, Bresnahan, Ha, Von Zastrow and Malenka2002; Huh et al., Reference Huh, Boulanger, Du, Riquelme, Brotz and Shatz2000; Paolicelli et al., Reference Paolicelli, Bolasco, Pagani, Maggi, Scianni, Panzanelli and Gross2011; Shatz, Reference Shatz2009; Stellwagen & Malenka, Reference Stellwagen and Malenka2006; Stephan, Barres, & Stevens, Reference Stephan, Barres and Stevens2012), myelination (Peferoen, Kipp, van der Valk, van Noort, & Amor, Reference Peferoen, Kipp, van der Valk, van Noort and Amor2014), and adult neurogenesis (Ekdahl, Claasen, Bonde, Kokaia, & Lindvall, Reference Ekdahl, Claasen, Bonde, Kokaia and Lindvall2003; Monje, Toda, & Palmer, Reference Monje, Toda and Palmer2003; Ziv et al., Reference Ziv, Ron, Butovsky, Landa, Sudai, Greenberg and Schwartz2006). Early exposure to inflammation has long-term impacts on the brain and behavior of rodents (Bilbo & Schwarz, Reference Bilbo and Schwarz2009; Knuesel et al., Reference Knuesel, Chicha, Britschgi, Schobel, Bodmer, Hellings and Prinssen2014; Patterson, Reference Patterson2009) and nonhuman primates (Short et al., Reference Short, Lubach, Karasin, Olsen, Styner, Knickmeyer and Coe2010). In humans, higher levels of serum inflammation in childhood has been associated with increased risk of depression and psychosis in adulthood (Khandaker et al., Reference Khandaker, Pearson, Zammit, Lewis and Jones2014).

The brain's specialized immune cells (e.g., microglia and T cells) and cytokines contribute to normal brain functioning and thus may be directly affected by disruptions in immune functioning (Boulanger, Reference Boulanger2009; Deverman & Patterson, Reference Deverman and Patterson2009; Yirmiya & Goshen, Reference Yirmiya and Goshen2011). The innate immune system supports tissue remodeling involved in mechanisms of brain plasticity. For example, microglia in their resting state play important roles in adult brain function (Hanisch & Kettenmann, Reference Hanisch and Kettenmann2007). T cells in the brain monitor and respond to signals in the cerebrospinal fluid and play a role in learning and memory in adults by stimulating neurogenesis and promoting the expression of brain-derived neurotrophic factor (BDNF) (Kipnis, Gadani, & Derecki, Reference Kipnis, Gadani and Derecki2012). Low levels of proinflammatory cytokines, produced by microglia and T cells, support brain plasticity (Yirmiya & Goshen, Reference Yirmiya and Goshen2011). Exaggerated or chronic neuroinflammation, however, is associated with impaired brain function such as poor learning and memory (Yirmiya & Goshen, Reference Yirmiya and Goshen2011). Microglia produce proinflammatory cytokines upon detection of inflammatory signals, prostaglandins, and free radicals, all of which can have neurotoxic effects and suppress neurogenesis (Hanisch & Kettenmann, Reference Hanisch and Kettenmann2007). Upon detection of inflammatory signals, T cells are involved in triggering protective immune responses that could be detrimental to brain function.

Third, inflammation affects microglial reactivity and can prime immune cells to be more reactive upon subsequent psychosocial stressors, resulting in increased inflammation. Early life inflammation can activate and prime microglia to subsequent inflammatory stimuli, thereby influencing later inflammatory responses to stressors (Perry & Holmes, Reference Perry and Holmes2014). Inflammation may downregulate neuronal ligands responsible for inhibiting this priming response (Perry & Holmes, Reference Perry and Holmes2014). Similar effects may occur if microglia are less sensitive to the inhibitory effect of glucocorticoids (Frank, Watkins, & Maier, Reference Frank, Watkins and Maier2013). Rodents with primed microglia exhibit increased proinflammatory cytokine production in the brain, increased neurotoxicity, and exaggerated sickness behavior when exposed to subsequent inflammatory stimulation (Perry & Holmes, Reference Perry and Holmes2014). Thus, microglia can be activated and primed by inflammatory stimuli and mount a greater inflammatory response from subsequent immune stimulation. Early life seems to be a sensitive period for this microglial priming (Bilbo & Schwarz, Reference Bilbo and Schwarz2009; Knuesel et al., Reference Knuesel, Chicha, Britschgi, Schobel, Bodmer, Hellings and Prinssen2014).

Fourth, inflammation can sensitize the neuroendocrine and immune systems in a bidirectional fashion (Danese & Lewis, Reference Danese and Lewis2017). In acute immune stimulation, the HPA axis is activated similar to activation following exposure to a stressor (Besedovsky, del Rey, Sorkin, & Dinarello, Reference Besedovsky, del Rey, Sorkin and Dinarello1986; Dunn, Reference Dunn2000). Evidence from animal models suggests that the immune system may program the HPA axis response during sensitive periods of development. In rodents, early life immune stimulation with LPS is associated with HPA axis abnormalities that are linked to psychopathology, including increased corticotropin-releasing hormone (CRH) levels, increased adrenocorticotropic hormone (ACTH) and corticosterone production in response to stress, decreased negative feedback sensitivity to glucocorticoids, and reductions in glucocorticoid receptor (GR) density in the brain (Pariante & Miller, Reference Pariante and Miller2001; Shanks, Larocque, & Meaney, Reference Shanks, Larocque and Meaney1995; Shanks et al., Reference Shanks, Windle, Perks, Harbuz, Jessop, Ingram and Lightman2000). If cytokines chronically stimulate the HPA axis, this can result in insufficient glucocorticoid signaling, inhibited BDNF secretion, inhibited nerve growth factor secretion, and inhibited cholinergic transmission (Dantzer et al., Reference Dantzer, O'Connor, Freund, Johnson and Kelley2008; Yirmiya & Goshen, Reference Yirmiya and Goshen2011). These results suggest that the immune system's programming of the HPA axis programming is part of the causal pathway between early life immune stimulation and psychopathology (Danese & Lewis, Reference Danese and Lewis2017). Together, the brain neurochemistry, developmental, priming, and neuroendocrine effects demonstrate evidence of direct and long-term risk for psychopathology (Boulanger, Reference Boulanger2009).

Early life adversity and inflammation

As ELA cannot be randomly assigned or manipulated in humans, animal models provide a support for causal inference in the link between early life stress and increased inflammation. Evidence from animal models point to associations between early life stress paradigms and immune system functioning (for comprehensive reviews, we point the reader to (Ganguly & Brenhouse, Reference Ganguly and Brenhouse2015; Hennessy, Deak, & Schiml-Webb, Reference Hennessy, Deak and Schiml-Webb2010a; Shanks & Lightman, Reference Shanks and Lightman2001). Although not analogous to all forms of ELA in humans, maternal separation as an ELA paradigm in nonhuman animal models paints the most consistent picture linking ELA with inflammation. Rodent and nonhuman primate models of maternal separation provide evidence for increased peripheral inflammatory biomarkers, markers of neuroinflammation, and greater proinflammatory response in response to subsequent stress (Coe, Rosenberg, & Levine, Reference Coe, Rosenberg and Levine1988; Cole et al., Reference Cole, Conti, Arevalo, Ruggiero, Heckman and Suomi2012; Hennessy, Deak, Schiml-Webb, Carlisle, & O'Brien, Reference Hennessy, Deak, Schiml-Webb, Carlisle and O'Brien2010b; Wieck, Andersen, & Brenhouse, Reference Wieck, Andersen and Brenhouse2013). Markers of neuroinflammation include blunted expression of proinflammatory mediators in the hippocampus (Wei, Simen, Mane, & Kaffman, Reference Wei, Simen, Mane and Kaffman2012), reduced microglial cell numbers in the midbrain areas of rodent pups (Chocyk et al., Reference Chocyk, Dudys, Przyborowska, Majcher, Mackowiak and Wedzony2011), increased expression of proinflammatory cytokine receptors in the hippocampus (Viviani et al., Reference Viviani, Boraso, Valero, Gardoni, Marco, Llorente and Viveros2014), changes in cortical microglial processes (Takatsuru, Nabekura, Ishikawa, Kohsaka, & Koibuchi, Reference Takatsuru, Nabekura, Ishikawa, Kohsaka and Koibuchi2015), and increased microglia activation (Brenhouse & Thompson, Reference Brenhouse and Thompson2015). Maternal separation in rodent models show a greater proinflammatory response in the brain and periphery when animals are exposed to later stressors (Avitsur, Hunzeker, & Sheridan, Reference Avitsur, Hunzeker and Sheridan2006; Brenhouse & Thompson, Reference Brenhouse and Thompson2015; Hennessy et al., Reference Hennessy, Deak, Schiml-Webb, Carlisle and O'Brien2010b).

Research Questions

Many thoughtful reviews, theoretical papers, and meta-analyses have been written on studies of ELA and inflammation in humans (Baumeister, Akhtar, Ciufolini, Pariante, & Mondelli, Reference Baumeister, Akhtar, Ciufolini, Pariante and Mondelli2016; Danese & Baldwin, Reference Danese and Baldwin2017; Danese & Lewis, Reference Danese and Lewis2017; Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020; Nusslock & Miller, Reference Nusslock and Miller2016). This domain of research is rapidly expanding, and this review brings together the outstanding and developmentally focused research questions moving forward. As the field has relatively heterogeneous inflammation outcome measures, we focus our discussion on CRP, IL-6, and tumor necrosis factor (TNF)-alpha, a common trio of inflammatory markers associated with psychopathology in humans (Yuan, Chen, Xia, Dai, & Liu, Reference Yuan, Chen, Xia, Dai and Liu2019).

We take a developmental focus because, in contrast to the studies on ELA and inflammation in adults, the literature on ELA and inflammation in infants, children, and adolescents is still in its early stages. A meta-analysis of 13 studies on ELA and inflammation in pediatric samples highlight the relative lack of studies overall, the high levels of heterogeneity of ELA and inflammation measures, and mixed results (Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020). Though this recent meta-analysis did not find a statistically significant association between ELA and inflammation, the authors argued that this was due to too much heterogeneity across pediatric studies (Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020). The ELA constructs, types of inflammation assessment, and timing of the inflammation assessment varied widely in these studies. The mixed results are not all together surprising when looking toward reviews of the animal literature, which find mixed results due to a multiplicity of stressors and inflammatory measures (Ganguly & Brenhouse, Reference Ganguly and Brenhouse2015; Hennessy et al., Reference Hennessy, Deak and Schiml-Webb2010a; Shanks & Lightman, Reference Shanks and Lightman2001). Therefore, as the field continues to grow, the field must address how to consistently measure ELA, consistently measure inflammation, test mediators, and account for moderators with a developmental lens. We refer the reader to Table 1, which codes the studies discussed relevant to the following research question sections.

Table 1. Studies of early life adversity and peripheral inflammation, arranged by mean age (years) at inflammation assessment

∅ = null association between ELA exposure and inflammatory outcome.

✓ = positive association between ELA exposure and inflammatory outcome.

BMI = body mass index; CPS = Child Protective Services; CRP = C-reactive protein; CTQ = Child Trauma Questionnaire; ELA = early life adversity; IL-6 = interleukin-6; MIDUS = Midlife in the United States; SES = socioeconomic status; TNF = tumor necrosis factor

Measuring early life stress

Are there features of ELA experiences or its measurement that are more strongly associated with inflammation? Within the study of ELA and inflammation in pediatric populations, we must clarify if there is a difference in inflammatory outcomes when measuring ELA prospectively versus retrospectively and when assessing ELA with objective or subjective measures. Further, would dimensional approaches to the measurement of ELA clarify research on ELA and inflammation if applied? For example, researchers have proposed that experiences of “deprivation” and “threat” in ELA contexts act as different dimensions and interact to produce a range of different outcomes (McLaughlin et al., Reference McLaughlin, Sheridan and Lambert2014). Finally, how does construct invariance in the measurement of ELA impact the research examining inflammatory outcomes longitudinally? Precision in the measurement and definition of ELA, then, is the first challenge if the field seeks to better understand the relationship between ELA, inflammation, and psychopathology across development.

Measurement of early life adversity

How adversity is assessed, whether through objective or self-report measures, prospectively or retrospectively, has long been a consideration in the study of child adversity. A recent study highlights this reporting discrepancy, as it found poor agreement between prospective and retrospective reports of childhood maltreatment within the same individuals (Baldwin, Reuben, Newbury, & Danese, Reference Baldwin, Reuben, Newbury and Danese2019). The literature provides no clarity about how the reporting of ELA affects inflammatory outcomes. Part of this lack of clarity arises from a lack of formal delineation between prospective measures and retrospective measures of ELA. Few studies on inflammation arising from ELA have prospective and retrospective measures to compare directly. One study that makes the comparison is the Dunedin Multidisciplinary Health and Development Study, which showed that inflammation at age 32 y was more strongly associated with prospective compared to retrospective measures of ELA (Reuben et al., Reference Reuben, Moffitt, Caspi, Belsky, Harrington, Schroeder and Danese2016). However, data from the 1958 British Cohort (N = 7,661) that included prospective and retrospective measures of victimization found more mixed results, with positive associations between ELA and inflammation dependent on the type of ELA exposure (Pinto Pereira et al., Reference Pinto Pereira, Stein Merkin, Seeman and Power2019).

Another related area that requires clarification is how the source of information on ELA influences outcomes. Retrospective measures are, by necessity, self-reports, while prospective measures can include objective information (e.g., court reports), parent reports, or self-reports on ELA. The few studies that do use objective measures of ELA found inconsistent evidence for a link between ELA and inflammation. Osborn and Widom (Reference Osborn and Widom2019) have recently shown that individuals with official reports of child maltreatment (specifically physical abuse) had significantly higher levels of CRP than nonmaltreated individuals, yet two studies using Child Protective Services (CPS) referrals found no association between CRP and maltreatment (Bernard et al., Reference Bernard, Hostinar and Dozier2019; Cicchetti et al., Reference Cicchetti, Handley and Rogosch2015). Other studies of adolescents and adults exposed to objectively and prospectively assessed ELA in the form of institutional care have found no association between neglect exposure and inflammatory biomarkers (Elwenspoek et al., Reference Elwenspoek, Hengesch, Leenen, Schritz, Sias, Schaan and Muller2017; Reid et al., Reference Reid, Coe, Doyle, Sheerar, Slukvina, Donzella and Gunnar2019; Slopen et al., Reference Slopen, Tang, Nelson, Zeanah, McDade, McLaughlin and Fox2019).

Perhaps the ultimate challenge across measures is that the effect sizes between ELA and inflammation are small, and without adequately powered studies the outcomes are predictably inconsistent. A meta-analysis of 25 studies on ELA and plasma levels of inflammation in adulthood found small but significant effect sizes, though most studies included ELA measured broadly and retrospectively in adulthood (Baumeister et al., Reference Baumeister, Akhtar, Ciufolini, Pariante and Mondelli2016). Prospective studies of ELA in large cohorts have also found positive associations between ELA and inflammation, though the significant inflammation biomarker outcome differs between them (Baldwin et al., Reference Baldwin, Arseneault, Caspi, Fisher, Moffitt, Odgers and Danese2018; Bucker et al., Reference Bucker, Fries, Kapczinski, Post, Yatham, Vianna and Kauer-Sant'Anna2015; Flouri, Francesconi, Midouhas, & Lewis, Reference Flouri, Francesconi, Midouhas and Lewis2020). Adequately powered studies in large, longitudinal cohorts with both prospective and retrospective measures of ELA and a consistent set of inflammation biomarkers could help clarify the issue of ELA measurement as it relates to inflammation risk.

Dimensional approaches to the measurement of early life stress type

A second question is whether inflammation outcomes differ depending on the dimension or type of adversity exposure. Numerous studies have found different outcomes when examining separate types of childhood adversity. In meta-analysis of ELA exposure and inflammation in pediatric populations, the types of ELA exposure were too different to compare dimensionally (Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020). Future research would benefit from outlining diversity in the type of adversity exposure to examine the development of a proinflammatory phenotype and risk for psychopathology. McLaughlin et al. (Reference McLaughlin, Sheridan and Lambert2014) propose a dimensional approach to disentangle the effects of ELA on neurodevelopment, and we propose that this dimensional approach could be applied to the study of inflammatory outcomes. The approach distinguishes “threat,” or experiences that threaten an individual's physical integrity, and “deprivation,” which encompasses the lack of species-expected environmental inputs and/or environmental complexity (McLaughlin et al., Reference McLaughlin, Sheridan and Lambert2014). While deprivation and threat often co-occur in contexts where children are victimized, measuring both may clarify differential inflammation and psychopathology outcomes. In this section, we review a few select studies to highlight the measurement questions that remain in this area.

Experiences of threat include those that involve physical or sexual violence, threat of death, or other physical harm (McLaughlin et al., Reference McLaughlin, Sheridan and Lambert2014). In adult populations, there are a few examples of examining “threat” types of childhood adversity as they relate to inflammatory outcomes. In subgroup analyses in a meta-analysis, the type of ELA exhibited contrasting results in specific inflammatory outcomes (Baumeister et al., Reference Baumeister, Akhtar, Ciufolini, Pariante and Mondelli2016). Childhood sexual abuse was positively associated with TNF-α but not with IL-6 or CRP. Childhood physical abuse was associated with TNF-α and IL-6 but not CRP (Baumeister et al., Reference Baumeister, Akhtar, Ciufolini, Pariante and Mondelli2016). An analysis of two large adult cohorts mentioned previously examined different types of childhood adversity on adult levels of inflammation (Pinto Pereira et al., Reference Pinto Pereira, Stein Merkin, Seeman and Power2019). In the British cohort, increased inflammation in adulthood was positively associated with physical abuse but not sexual abuse (Pinto Pereira et al., Reference Pinto Pereira, Stein Merkin, Seeman and Power2019). In the US cohort, physical abuse was not associated with either IL-6 or CRP in adulthood (Pinto Pereira et al., Reference Pinto Pereira, Stein Merkin, Seeman and Power2019). However, childhood sexual abuse was significantly associated with IL-6 and CRP for nonwhites only (Pinto Pereira et al., Reference Pinto Pereira, Stein Merkin, Seeman and Power2019). In studies of pediatric populations, threat experiences of maltreatment and victimization are positively associated with inflammation in some studies (Baldwin et al., Reference Baldwin, Arseneault, Caspi, Fisher, Moffitt, Odgers and Danese2018; Bucker et al., Reference Bucker, Fries, Kapczinski, Post, Yatham, Vianna and Kauer-Sant'Anna2015; Danese et al., Reference Danese, Pariante, Caspi, Taylor and Poulton2007; Flouri et al., Reference Flouri, Francesconi, Papachristou, Midouhas and Lewis2019; Kilic et al., Reference Kilic, Ozlem and Murat2017) but not all (Bernard et al., Reference Bernard, Hostinar and Dozier2019; Cicchetti et al., Reference Cicchetti, Handley and Rogosch2015; Miller & Cole, Reference Miller and Cole2012).

One component of the dimensional approach to measuring ELA is the grounding of the dimensions in current evidence on neurodevelopment (McLaughlin et al., Reference McLaughlin, Sheridan and Lambert2014). Threat experiences may result in changes to emotional learning networks (Johansen, Cain, Ostroff, & LeDoux, Reference Johansen, Cain, Ostroff and LeDoux2011; LeDoux, Reference LeDoux2003; McLaughlin et al., Reference McLaughlin, Sheridan and Lambert2014). Kraynak et al. looked at pathways from childhood abuse to systemic inflammation and corticolimbic functionality in healthy adults and found that childhood abuse was associated with increased IL-6 and indirectly associated with reduced amygdala– ventromedial prefrontal cortex (vmPFC) connectivity (2019). Consistent with recent neurobiological models of early life threat experiences, associations between childhood physical abuse and adulthood corticolimbic circuit functionality may be partially explained by inflammatory processes (Kraynak et al., Reference Kraynak, Marsland, Hanson and Gianaros2019). Interestingly, while CRP associated negatively with amygdala– ventromedial prefrontal cortex connectivity, it did not statistically associate with childhood physical abuse (Kraynak et al., Reference Kraynak, Marsland, Hanson and Gianaros2019). This study provides preliminary evidence of how a dimensional and neurobiological approach may elucidate ELA–inflammation–psychopathology connections.

In contrast to the construct of “threat,” deprivation experiences are defined as the absence of expected environmental inputs (i.e., cognitive inputs, social inputs, and environmental stimuli that are species- and age-typical in their complexity (McLaughlin et al., Reference McLaughlin, Sheridan and Lambert2014). Experiences of deprivation are central for children who have been institutionalized (i.e., orphanage care), children experiencing neglect, and children in contexts of poverty. Few studies of adult cohorts operationalize neglect as a separate dimensional construct, and the definition of neglect is heterogeneous across and within these studies. For example, emotional neglect was not associated with either IL-6 or CRP in a large US cohort and physical neglect but not emotional neglect was positively associated with inflammation in a large British cohort (Pinto Pereira et al., Reference Pinto Pereira, Stein Merkin, Seeman and Power2019). The Baumeister meta-analysis demonstrated that parental absence was significantly and positively associated with CRP, though the measures of deprivation across the studies were highly inconsistent (Baumeister et al., Reference Baumeister, Akhtar, Ciufolini, Pariante and Mondelli2016).

Studies of adults and children who experienced institutional care early in life provide an opportunity to examine the dimension of deprivation as it relates to inflammatory outcomes. Institutional care typically involves environmental deprivation, caregiver separation, and neglect. Two studies that focused on neglect exposure and subsequent inflammation compared previously institutionalized (PI) adolescents to community controls. One study examined youth adopted out of institutional care before the age of 2 y into well-resourced families in the United States (Reid et al., Reference Reid, Coe, Doyle, Sheerar, Slukvina, Donzella and Gunnar2019) and another examined youth who either remained in institutional care or were randomly assigned to a foster care intervention (Slopen et al., Reference Slopen, Tang, Nelson, Zeanah, McDade, McLaughlin and Fox2019). These studies of adolescents removed from institutional care as infants provide opportunities to look at sensitive periods of adversity exposure as it relates to inflammation and compare differences in behavioral and environmental inputs. Neither study found evidence of increased peripheral inflammation in PI adolescents (Reid et al., Reference Reid, Coe, Doyle, Sheerar, Slukvina, Donzella and Gunnar2019; Slopen et al., Reference Slopen, Tang, Nelson, Zeanah, McDade, McLaughlin and Fox2019). One study found no differences in plasma levels of inflammation in PI and comparison adolescents (Reid et al., Reference Reid, Coe, Doyle, Sheerar, Slukvina, Donzella and Gunnar2019), which replicated findings in PI adults (Elwenspoek et al., Reference Elwenspoek, Hengesch, Leenen, Schritz, Sias, Schaan and Muller2017a). These studies are consistent with a study with the Bucharest Early Intervention Project, in which inflammation (CRP, IL-6, IL-8, TNF-α) was measured in dried blood spots in adolescent follow-up of institutionalized infants randomized to care as usual (n = 68) or foster care intervention (n = 68), and compared to never institutionalized controls (n = 127). The study found no differences in levels of peripheral inflammation markers between the PI and comparison adolescents (Slopen et al., Reference Slopen, Tang, Nelson, Zeanah, McDade, McLaughlin and Fox2019). However, further post hoc analyses found differences in CRP only in PI adolescents who exhibited an increasing adiposity trajectory and glucose dysregulation, though this group of adolescents was a small subset of the full PI sample (Tang et al., Reference Tang, Slopen, Nelson, Zeanah, Georgieff and Fox2018).

Unfortunately, none of the studies reviewed have a dimensional measure of threat and deprivation, and many of the conditions of threat and deprivation co-occur. Current studies suggest that inflammatory outcomes can be diverse even between different threat and deprivation exposures. The field would be well served by research that includes dimensional measures of threat and deprivation in better understand how dimensional constructs of ELA do – or do not – result in a proinflammatory phenotype.

Construct invariance from infancy to childhood and adolescence

Longitudinal construct invariance is the third measurement research question for the study of ELA and inflammation. Many studies measure the family environment in infancy and early childhood, as this proximal family environment is thought to best capture experiences of adversity at very young ages. David, Measelle, Ostlund, and Ablow (Reference David, Measelle, Ostlund and Ablow2017) found that salivary CRP was positively and independently associated with socioeconomic disadvantage and maternal stress in 18-month-olds. In contrast, a study of 125 5-year-olds found no association between family level adversity and salivary IL-6 or TNF-alpha (Riis et al., Reference Riis, Granger, Minkovitz, Bandeen-Roche, DiPietro and Johnson2016). In a prospective, longitudinal cohort of 600 adolescents, exposure to family-level adversity in infancy predicted higher levels of CRP, mediated through higher BMI in childhood (Reid et al., Reference Reid, Doom, Argote, Correa-Burrows, Lozoff, Blanco and Gahagan2020). Finally, a study of 337 11-year-olds found no association between family-level adversity in infancy and IL-6 or TNF-alpha. The study found significant associations only between caregiver depressive symptoms in infancy and higher levels of CRP in childhood (O'Connor et al., Reference O'Connor, Willoughby, Moynihan, Messing, Vallejo Sefair, Carnahan and Caserta2020). Few studies examine ELA in infancy and early childhood, and none yet provide clarity around construct invariance from family adversity to later forms of ELA and inflammation.

Measuring inflammation/immune biomarkers

Questions on the measurement of inflammation are equally important to the questions of measuring ELA. Namely, what are the best inflammation biomarkers to measure associations between ELA, inflammation, and psychopathology? The nature of human research on psychopathology is that we are limited primarily to inflammatory biomarkers in the periphery, not the brain. This some challenges, which include (a) the interpretation of peripheral blood biomarkers of inflammation to brain measures of inflammation (i.e., tissue specificity) and (b) the selection of inflammation biomarkers from many different inflammatory cytokine options.

Tissue specificity

Though the connection between peripheral inflammation and neuroinflammation has been made (Miller & Raison, Reference Miller and Raison2016), there are still open questions that researchers should continue to acknowledge when studying adversity, inflammation, and psychopathology risk. A meta-analysis of studies examining cerebrospinal fluid, positron emission tomography and postmortem brain tissue in patients with depression found that patients with depression have higher cerebral spinal fluid levels of IL-6 and TNF-α than controls. However, there was little correlation between central and peripheral markers of inflammation in the studies reviewed, which may reflect a lack of connection between central and peripheral inflammation or other possible moderating factors (Enache, Pariante, & Mondelli, Reference Enache, Pariante and Mondelli2019). Nonetheless, measures in blood, and therefore blood draws, are considered the gold standard for inflammatory biomarker measurement in population-based studies. Most studies investigating the links between ELA and inflammation focus on blood as the target tissue of interest in populations of adults, adolescents, and many studies of children.

Large-scale studies and studies of infants and children often use less invasive biomarkers for acceptability and cost. Blood draws can be cost-prohibitive, not feasible, and are not minimally invasive. One less invasive methods of collection for inflammatory biomarkers involve finger prick and blood spot (McDade, Williams, & Snodgrass, Reference McDade, Williams and Snodgrass2007). The blood spot cards are let to dry and can be stored until laboratory analysis with good stability. Levels of inflammation biomarkers, such as CRP, in dried blood spot correlate highly with levels in serum (r = 0.96) (McDade, Burhop, & Dohnal, Reference McDade, Burhop and Dohnal2004). Analysis of ELA-related inflammation abnormalities in dried blood spot showed results comparable to those in serum (Copeland et al., Reference Copeland, Wolke, Lereya, Shanahan, Worthman and Costello2014; Danese et al., Reference Danese, Caspi, Williams, Ambler, Sugden, Mika and Arseneault2011). As such, there is interest in developing further inflammation biomarkers including cytokines from dried blood spots.

Blood draws and dried blood spots may be considered too invasive in studies with healthy infants or young children. Reflecting this, the majority of the studies in infancy and early childhood (0–5 years) in this review use salivary markers of inflammation, which limits our ability to compare these with studies in older children and adults. Several studies have reported poor correlations between saliva and blood for inflammatory markers (Dillon et al., Reference Dillon, Holmes, Birk, Brooks, Lyons-Ruth and Pizzagalli2009; Riis et al., Reference Riis, Granger, Minkovitz, Bandeen-Roche, DiPietro and Johnson2016), likely due to the relative difficulty that most proteins have in passing the multiple barriers between blood and saliva compartments. Cytokines are too large to enter the mouth from the periphery through passive diffusion and instead enter through sites of inflammation or tissue damage, producing large variability between individuals (Bosch, Reference Bosch2014). Thus, while salivary cytokines are mostly indicative of local inflammation (Bosch, Reference Bosch2014), they may still serve as important indicators of health status while taking care to control for dental health confounds (Slavish, Graham-Engeland, Smyth, & Engeland, Reference Slavish, Graham-Engeland, Smyth and Engeland2015). As in any tissue, the type of inflammatory biomarker assessed is important. CRP from saliva has been associated with CRP in circulating blood more consistently than other inflammatory biomarkers, perhaps because CRP is produced in the liver, not in the mouth, and its most likely route into saliva is via blood (Slavish et al., Reference Slavish, Graham-Engeland, Smyth and Engeland2015). A validation study of a high-sensitivity CRP assay in saliva allowed prediction of serum CRP in healthy adults (Ouellet-Morin, Danese, Williams, & Arseneault, Reference Ouellet-Morin, Danese, Williams and Arseneault2011). Markers of salivary inflammation have been stable across time in adolescent samples in the short and long term (when averaging across multiple assessments) (Shields, Slavich, Perlman, Klein, & Kotov, Reference Shields, Slavich, Perlman, Klein and Kotov2019). More research is needed to establish both the health relevance and the mechanisms driving salivary inflammation generally and across development. Further research is needed to determine how ELA relates to salivary inflammation, what increased salivary inflammation means for infants and young children in the short term, whether this measure is associated with blood levels of inflammation, or what the long-term implications of increased salivary inflammation are.

Biomarker (cytokine) selection

Diverse inflammatory markers have been used to test the association between ELA and inflammation. To counteract the literature's heterogeneity, longitudinal and system-wide studies of inflammation regulation with multiple biomarkers that can differentiate trait- and state-specific changes in the context of childhood adversity would improve our understanding of etiology and establish valuable biomarkers. Del Guidice and Gangestad make the argument that IL-6 and CRP assays alone are not enough to establish the existence of inflammatory states due to their multiple roles and states in the body, a concept accepted in immunology research but rarely addressed in behavioral research (Del Giudice & Gangestad, Reference Del Giudice and Gangestad2018). For example, IL-6 and CRP might be elevated for maintenance functions and not necessarily indicative of an aberrant and chronic inflammatory state. Future studies should consider using IL-6 and CRP alongside additional inflammatory biomarkers (e.g., IL-1B, TNF-alpha, and circulating levels of soluble IL-6 receptors such as sIL-6R) (Del Giudice & Gangestad, Reference Del Giudice and Gangestad2018). A meta-analysis of 704 publications on psychopathology and 44 inflammatory biomarkers found that related disorders share similar patterns of inflammatory changes (Yuan et al., Reference Yuan, Chen, Xia, Dai and Liu2019). Longitudinal studies of inflammation regulation with multiple biomarkers could help differentiate trait- and state-specific changes in psychopathology to establish valuable biomarkers in the ELA literature (Yuan et al., Reference Yuan, Chen, Xia, Dai and Liu2019).

Mediation: Mechanisms linking early life stress and inflammation

Mechanisms through which ELA could contribute to chronic inflammation include genetic risk, exposure to acute immune responses early in life, behavior, neuroendocrine dysregulation, adipose tissue, gut microbiome dysbiosis, and stress-induced disruptions to nutrient metabolism, among others. This review is not exhaustive but seeks to highlight important and modifiable pathways by which ELA could contribute to inflammation and increase the subsequent risk for psychopathology. We integrate insights from provide support for the current theoretical mechanisms and offer alternative or contradicting evidence to inform research agendas going forward.

Gene-environment correlation

The genetic vulnerability of inflammation is estimated to account for between 13%–30% of within-subjects variability of CRP (Pankow et al., Reference Pankow, Folsom, Cushman, Borecki, Hopkins, Eckfeldt and Tracy2001), and there may be shared, underlying genetic vulnerability to ELA, psychopathology, and inflammation (2017). Immune-related genetic pathways have been found to predict risk for several types of psychopathology (Network & Pathway Analysis Subgroup of Psychiatric Genomics, 2015). Danese & Baldwin (Reference Danese and Baldwin2017) note that an individual exhibiting early behavioral expressions of genetic vulnerability to psychopathology (e.g., emotional dysregulation, oppositional behavior) might increase their risk of maltreatment in childhood. However, the study on victimization by Baldwin et al. (Reference Baldwin, Arseneault, Caspi, Fisher, Moffitt, Odgers and Danese2018) with a genetically informative sample of twins found that exposure to ELA was associated with increased inflammation above and beyond the genetic risk for increased inflammation, further suggesting that while genetics plays a role, additional mechanisms are at play.

Acute inflammatory responses

Acute inflammation in response to physical trauma, infection, or exposure to pathogens during sensitive periods early in life may stimulate microglia and neuroendocrine activity and affect brain development. Acute inflammatory responses can be incited through psychological stress or in the context of physical trauma, as children exposed to ELA are more likely to be exposed to physical injuries (Gilbert et al., Reference Gilbert, Widom, Browne, Fergusson, Webb and Janson2009). Children exposed to ELA are more likely to be exposed to infection (Gilbert et al., Reference Gilbert, Widom, Browne, Fergusson, Webb and Janson2009) and psychological stress and traumatic experiences can increase susceptibility to both infections and recurrent viral infections (Cohen et al., Reference Cohen, Janicki-Deverts, Doyle, Miller, Frank, Rabin and Turner2012; Shirtcliff, Coe, & Pollak, Reference Shirtcliff, Coe and Pollak2009). ELA could contribute to systemic inflammation through these recurrent acute immune perturbations, but this pathway may not be applicable in all contexts. For example, in the studies of PI adolescents and young adults mentioned earlier in this paper, ELA-exposed youth have been found to exhibit higher levels of cytomegalovirus (CMV) titers, likely due to increased exposure to infectious disease and pathogens in institutional care (Elwenspoek et al., Reference Elwenspoek, Sias, Hengesch, Schaan, Leenen, Adams and Turner2017b; Reid et al., Reference Reid, Coe, Doyle, Sheerar, Slukvina, Donzella and Gunnar2019). ELA and CMV exposure were associated with acquired immune system perturbations and markers of T-cell senescence, but not higher levels of inflammation (Elwenspoek et al., Reference Elwenspoek, Sias, Hengesch, Schaan, Leenen, Adams and Turner2017b; Reid et al., Reference Reid, Coe, Doyle, Sheerar, Slukvina, Donzella and Gunnar2019). In addition, McDade's study of a prospective birth cohort in the Philippines found that ELA was associated with adult levels of inflammation only in individuals with low levels of microbial exposure in infancy (McDade, Rutherford, Adair, & Kuzawa, Reference McDade, Rutherford, Adair and Kuzawa2010). It is unclear how ELA and early infection interact, the salience of specific pathogen exposures, and how infection affects the developing immune system. For instance, PI youth are not at increased risk of more inflammation (Elwenspoek et al., Reference Elwenspoek, Hengesch, Leenen, Schritz, Sias, Schaan and Muller2017a; Reid et al., Reference Reid, Coe, Doyle, Sheerar, Slukvina, Donzella and Gunnar2019; Slopen et al., Reference Slopen, Tang, Nelson, Zeanah, McDade, McLaughlin and Fox2019), but they are at increased risk of psychopathology (Gunnar & Reid, Reference Gunnar and Reid2019). In the Philippines cohort, there was no evidence of association between depressive symptomology in adulthood and increased inflammation (McDade, Borja, Adair, & Kuzawa, Reference McDade, Borja, Adair and Kuzawa2013). In contexts where infectious disease burden is high, pathogen exposure may act as a protective factor in the pathway from ELA, inflammation, and psychopathology (McDade, Reference McDade2012). Future work should consider operationalizing pathogen and infection exposure across development to better understand inflammation and psychopathology risk.

Neuroendocrine dysregulation

Psychological stress can indirectly contribute to inflammation by inducing neuroendocrine abnormalities and changes in HPA axis physiology. The HPA axis is a key neuroendocrine pathway involved in processes critical to homeostasis, including an organism's responses to challenge, metabolism, and immune activity. Peripheral and central HPA axis mediators affect brain function (McEwen, Weiss, & Schwartz, Reference McEwen, Weiss and Schwartz1968), and early life experiences can significantly shape the development and subsequent functioning of the HPA axis (Levine, Alpert, & Lewis, Reference Levine, Alpert and Lewis1957). Furthermore, the HPA axis and the immune system relate to each other in dynamic and bidirectional ways. Inflammation can activate the HPA axis and cortisol activates anti-inflammatory and suppresses proinflammatory responses (Barnes & Adcock, Reference Barnes and Adcock2009; Besedovsky et al., Reference Besedovsky, del Rey, Sorkin and Dinarello1986). In situations where chronic stress leads to HPA upregulation, immune cells can become insensitive to the GR-mediated inhibitory effects of cortisol after excessive exposure over long periods, thought to be indicative of “glucocorticoid resistance” (Cohen et al., Reference Cohen, Janicki-Deverts, Doyle, Miller, Frank, Rabin and Turner2012; Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020; Miller, Chen, & Parker, Reference Miller, Chen and Parker2011b).

HPA biology in humans is typically measured in the context of cortisol reactivity to stress and the diurnal rhythm of cortisol secretion, which reflect the ability of the HPA axis to regulate responses to stress and to modulate circadian rhythm. In chronic ELA exposures, the evidence suggests that the HPA axis responsivity can either upregulate, where cortisol becomes chronically elevated, or downregulate, where the HPA axis becomes become flat and unresponsive (McEwen, Reference McEwen2007, Reference McEwen2012). In general, current evidence shows that childhood adversity is associated with heightened activity of the axis (Koss & Gunnar, Reference Koss and Gunnar2018; Sanchez et al., Reference Sanchez, Ladd and Plotsky2001). If the axis is chronically elevated, it may produce a blunted cortisol response (Koss & Gunnar, Reference Koss and Gunnar2018; Sanchez et al., Reference Sanchez, Ladd and Plotsky2001).

Blunted cortisol patterns have been found in studies of PI children (Koss, Hostinar, Donzella, & Gunnar, Reference Koss, Hostinar, Donzella and Gunnar2014; Koss, Mliner, Donzella, & Gunnar, Reference Koss, Mliner, Donzella and Gunnar2016) and in children experiencing neglect and maltreatment (Bernard, Hostinar, & Dozier, Reference Bernard, Hostinar and Dozier2015; Bruce, Fisher, Pears, & Levine, Reference Bruce, Fisher, Pears and Levine2009). Blunted cortisol patterns may lead to different Neuroendocrine×Immune System interactions. Inflammatory cytokine production is typically inhibited when cortisol binds to GRs in healthy immune cells (Barnes & Adcock, Reference Barnes and Adcock2009). Thus, in the case of downregulated HPA activity, low circulating levels of cortisol might induce elevated levels of inflammatory cytokines (Del Giudice & Gangestad, Reference Del Giudice and Gangestad2018). Studies have found insufficient glucocorticoid signaling in adolescents and adults with a history of childhood maltreatment, which could bring about systemic inflammation (Heim, Ehlert, & Hellhammer, Reference Heim, Ehlert and Hellhammer2000; Miller, Cohen, & Ritchey, Reference Miller, Cohen and Ritchey2002; Miller, Freedland, Carney, Stetler, & Banks, Reference Miller, Freedland, Carney, Stetler and Banks2003). However, this finding is not always consistent: the study of 11-year-olds mentioned previously found no association between glucocorticoid resistance and either ELA exposure in infancy or inflammation at 11 years, even though early adversity exposure was positively associated with inflammation (O'Connor et al., Reference O'Connor, Willoughby, Moynihan, Messing, Vallejo Sefair, Carnahan and Caserta2020). It is unclear whether this discrepancy is a result of the need for cumulative adversities to result in glucocorticoid resistance, a level of adversity that was not severe enough to impact glucocorticoid resistance, or an indication of multiple adversity-to-inflammation pathways across development.

Proinflammatory effects on immune cells in development may only be reliably observed when immune cells are stimulated in vitro, not when measured in circulation (Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020). If youth exposed to ELA exhibit elevated concentrations of cortisol, this upregulation may be masking the proinflammatory phenotype in children if inflammation is measured with circulating markers (Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020; Tarullo & Gunnar, Reference Tarullo and Gunnar2006). Chronic HPA activation during sensitive developmental periods can induce epigenetic changes in the GR, reducing signaling in a compensatory manner (Weaver et al., Reference Weaver, Cervoni, Champagne, D'Alessio, Sharma, Seckl and Meaney2004). This signal reduction could lead to resistance to cortisol's anti-inflammatory properties. ELA is associated with DNA demethylation in the glucocorticoid response elements of the FK506 binding protein 5 (FKBP5) gene and reduced sensitivity to glucocorticoids’ inhibitory effect on IL-6 production in peripheral blood immune cells stimulated in vitro (Klengel et al., Reference Klengel, Mehta, Anacker, Rex-Haffner, Pruessner, Pariante and Binder2013). A study in 18-year-old females retrospectively reporting ELA did not find higher levels of peripheral IL-6, but in vitro analyses found pronounced cytokine responses to bacterial challenge and evidence for decreased efficacy of cortisol to regulate the inflammatory response (Miller & Chen, Reference Miller and Chen2010). HPA axis and immune system interactions highlight the need to consider how neuroendocrine responses to adversity may increase risk for psychopathology through alterations in the co-regulation of multiple systems. Few studies on childhood adversity investigate HPA axis functioning with immune functioning (Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020). More complex models might clarify how childhood adversity impacts the brain and behavior through both HPA- and immune-mediating systems.

Gut microbiome dysbiosis

Growing evidence suggests that the microbiome plays a role in the pathway between ELA, stress-mediating and immune-system development, and mental health. The gut microbiome is involved in the immune system development and function (Hooper, Littman, & Macpherson, Reference Hooper, Littman and Macpherson2012). Its effects include changes in inflammation (Cani, Osto, Geurts, & Everard, Reference Cani, Osto, Geurts and Everard2012; Wells et al., Reference Wells, Brummer, Derrien, MacDonald, Troost, Cani and Garcia-Rodenas2017) and immune competence in response to pathogen exposure and infection (Mackos, Maltz, & Bailey, Reference Mackos, Maltz and Bailey2017; Zanella Terrier, Simonet, Bichard, & Frossard, Reference Zanella Terrier, Simonet, Bichard and Frossard2014). Gut microbiome dysbiosis has been associated with psychological impairments (Cryan & Dinan, Reference Cryan and Dinan2012; McVey Neufeld, Mao, Bienenstock, Foster, & Kunze, Reference McVey Neufeld, Mao, Bienenstock, Foster and Kunze2013). ELA may alter communication between the gut and the brain through altered vagus nerve and enteric nervous system signaling, HPA axis activation, immunomodulation, and inflammation (Cryan & Dinan, Reference Cryan and Dinan2012; Gareau, Silva, & Perdue, Reference Gareau, Silva and Perdue2008; Konturek, Brzozowski, & Konturek, Reference Konturek, Brzozowski and Konturek2011; Segerstrom & Miller, Reference Segerstrom and Miller2004). Psychosocial stress has been associated with changes in gut microbiota structure and activity (Mackos et al., Reference Mackos, Maltz and Bailey2017). In rodent and nonhuman primate models, early maternal separation has transient and long-term effects on the gut microbiota (Bailey & Coe, Reference Bailey and Coe1999; O'Mahony et al., Reference O'Mahony, Marchesi, Scully, Codling, Ceolho, Quigley and Dinan2009). In murine models, ELS-induced alterations of the microbiome persist into adulthood (Garcia-Rodenas et al., Reference Garcia-Rodenas, Bergonzelli, Nutten, Schumann, Cherbut, Turini and Corthesy-Theulaz2006; Jasarevic, Howard, Misic, Beiting, & Bale, Reference Jasarevic, Howard, Misic, Beiting and Bale2017; Jasarevic, Rodgers, & Bale, Reference Jasarevic, Rodgers and Bale2015). Changes to the gut microbiome in these models have been associated with anxiety-like behaviors and activation of stress-mediating systems (Gareau et al., Reference Gareau, Silva and Perdue2008). Rodents exposed to stress exhibit inflammation, altered gastrointestinal (GI) function and permeability, and differences in immune activity (Gareau et al., Reference Gareau, Silva and Perdue2008). In humans, differences in the gut microbiome have been found in adults who have developed PTSD after trauma exposure and adults with a history of childhood trauma (Hemmings et al., Reference Hemmings, Malan-Muller, van den Heuvel, Demmitt, Stanislawski, Smith and Lowry2017). ELA-exposed children have also been found to exhibit gut microbiome differences (Callaghan et al., Reference Callaghan, Fields, Gee, Gabard-Durnam, Caldera, Humphreys and Tottenham2020). Although conducted in a small sample of children, this study observed that gut bacteria levels were correlated with prefrontal cortex activation to emotional faces (Callaghan et al., Reference Callaghan, Fields, Gee, Gabard-Durnam, Caldera, Humphreys and Tottenham2020). The relationship between ELA-induced gut dysbiosis and inflammation are likely bidirectional (Cryan & Dinan, Reference Cryan and Dinan2012; Hooper et al., Reference Hooper, Littman and Macpherson2012).

Stress-induced nutrient metabolism disruptions

Disruptions to nutrient metabolism is an unexplored pathway by which ELS can lead to inflammation. To function properly, the developing immune system requires macronutrients (i.e., protein, fat, carbohydrates) and micronutrients (e.g., iron, zinc, B vitamins etc.). Psychosocial stress and dysregulated neuroendocrine pathways can disrupt nutrient metabolism even in the context of adequate nutrient intake (Monk, Georgieff, & Osterholm, Reference Monk, Georgieff and Osterholm2013; Suchdev et al., Reference Suchdev, Boivin, Forsyth, Georgieff, Guerrant and Nelson2017) and lead to through disruptions in immune functioning. There are many required nutrients for immune system development, but we will use iron as an illustrative example. Iron deficiency is one of the most common forms of malnutrition, with an estimated 4 in 10 children under 5 being affected (Stevens et al., Reference Stevens, Finucane, De-Regil, Paciorek, Flaxman and Branca2013), and children exposed to ELA are often at higher risk of iron deficiency. Adequate iron intake and utilization is necessary for normal white blood cell and cytokine function (Coe, Lubach, & Shirtcliff, Reference Coe, Lubach and Shirtcliff2007). In murine models, early iron deficiency increased expression of proinflammatory cytokine gene pathways in the adult brain even after iron repletion. Neonatal iron deficiency also reduced anti-inflammatory gene expression and altered cell-mediated immune response genes in infant mice (Tran, Singh, Wallin, & Georgieff, Reference Tran, Singh, Wallin and Georgieff2018). Research from a nonhuman primates showed that exposure to early life stress resulted in lower levels of iron and iron status subsequently predicted greater deficits in innate immune system functioning (Coe et al., Reference Coe, Lubach and Shirtcliff2007). Few studies exist in human models, but one study found that a randomized control trial of iron supplementation was associated with decreased levels of inflammation in a cohort of adolescents, even after exposure to ELA in infancy (Reid et al., Reference Reid, Doom, Argote, Correa-Burrows, Lozoff, Blanco and Gahagan2020). ELA-induced nutrient dysregulation could be a mediator that is ripe for research and intervention.

Adipose tissue

Adipose tissue generates and regulates about one-third of circulating inflammatory markers (Black, Reference Black2003). Obesity in childhood is associated with low-grade inflammation (Visser, Bouter, McQuillan, Wener, & Harris, Reference Visser, Bouter, McQuillan, Wener and Harris2001), and ELA is a risk factor for obesity in experimental animal models and human studies (Bzostek & Beck, Reference Bzostek and Beck2011; Danese & Tan, Reference Danese and Tan2014; Gundersen, Mahatmya, Garasky, & Lohman, Reference Gundersen, Mahatmya, Garasky and Lohman2011; Jelleyman & Spencer, Reference Jelleyman and Spencer2008; Weinberg et al., Reference Weinberg, Dietz, Stoyak, Melhem, Porta, Payne and Brent2013). The links between ELA and increased adiposity could arise from a biologically and behaviorally driven “thrifty” phenotype characterized by increased energy intake, increased storage, and/or decreased energy expenditure (Danese & Tan, Reference Danese and Tan2014). Childhood stress has been linked with emotional eating and unhealthy dietary patterns (Michels et al., Reference Michels, Sioen, Braet, Eiben, Hebestreit, Huybrechts and De Henauw2012), perhaps due to “self-medicating” with energy dense foods (Baldwin & Danese, Reference Baldwin and Danese2019). Impaired reward processing and reduced self-control from ELA may lead to increased intake of high-fat, high-sugar foods (Evans, Fuller-Rowell, & Doan, Reference Evans, Fuller-Rowell and Doan2012; Hostinar, Ross, Chen, & Miller, Reference Hostinar, Ross, Chen and Miller2015b). The neuroendocrine pathway between ELA, obesity, and inflammation could be a potent one, as glucocorticoids contribute to adipose tissue development (Dallman et al., Reference Dallman, Strack, Akana, Bradbury, Hanson, Scribner and Smith1993) and cortisol plays a central role in regulating food intake and metabolism (Dallman, Reference Dallman2010; Dallman, Pecoraro, & la Fleur, Reference Dallman, Pecoraro and la Fleur2005). Thus, early adversity exposure could increase the risk for children and adolescents to engage in eating behaviors that contribute to increased adiposity and inflammation.

ELA could result in obesity through decreased energy expenditure from metabolic hormone alterations and mental health problems (Baldwin & Danese, Reference Baldwin and Danese2019). ELA is associated with changes in leptin levels, which are responsible for lipolysis (Danese et al., Reference Danese, Dove, Belsky, Henchy, Williams, Ambler and Arseneault2014; Panagiotaropoulos et al., Reference Panagiotaropoulos, Papaioannou, Pondiki, Prokopiou, Stylianopoulou and Gerozissis2004). Mental health problems associated with child adversity are also associated with physical inactivity (Baldwin & Danese, Reference Baldwin and Danese2019). Impaired functioning in neuroendocrine and leptin pathways could dysregulate thermogenesis and lipolysis, leading to decreased energy expenditure and increased adipose tissue growth (Danese & Tan, Reference Danese and Tan2014). Inflammation itself can induce fatigue (Dantzer et al., Reference Dantzer, O'Connor, Freund, Johnson and Kelley2008), and thus inflammation from ELA exposure could cyclically contribute to reduced energy expenditure, increased adipose tissue, and increased inflammation.

Studies have found that ELA was associated with adolescent levels of inflammation only in the context of accelerated BMI growth trajectories (Reid et al., Reference Reid, Doom, Argote, Correa-Burrows, Lozoff, Blanco and Gahagan2020; Tang et al., Reference Tang, Slopen, Nelson, Zeanah, Georgieff and Fox2018). In a number of studies, adiposity attenuated or abolished several of the associations between ELA and inflammation (Matthews et al., Reference Matthews, Chang, Thurston and Bromberger2014; Pinto Pereira et al., Reference Pinto Pereira, Stein Merkin, Seeman and Power2019), though others have found that the association between adversity and inflammation was independent of obesity in both adult and pediatric samples (Baldwin et al., Reference Baldwin, Arseneault, Caspi, Fisher, Moffitt, Odgers and Danese2018; Danese et al., Reference Danese, Caspi, Williams, Ambler, Sugden, Mika and Arseneault2011; Danese et al., Reference Danese, Pariante, Caspi, Taylor and Poulton2007; O'Connor et al., Reference O'Connor, Willoughby, Moynihan, Messing, Vallejo Sefair, Carnahan and Caserta2020). This adversity–obesity–inflammation pathway seems to be of increased importance in studies of childhood adversity and inflammation in adolescence.

Brain functioning and behavior

ELA can instantiate a chronic inflammatory state through brain and behavioral changes. The Neuroimmune Network Hypothesis outlines the process by which ELA prompts crosstalk between the neural and immune systems to create a loop of increased threat sensitivity, inflammation, and unhealthy behaviors that can result in increased risk for chronic inflammation (Nusslock & Miller, Reference Nusslock and Miller2016). ELA is associated with impaired executive function, impaired reward processing, and hippocampal and prefrontal cortex abnormalities in human and nonhuman animal studies, which could lead to impaired ability to suppress unwanted behavior (Brunson et al., Reference Brunson, Kramar, Lin, Chen, Colgin, Yanagihara and Baram2005; Danese & McEwen, Reference Danese and McEwen2012; Dillon et al., Reference Dillon, Holmes, Birk, Brooks, Lyons-Ruth and Pizzagalli2009; Matthews & Robbins, Reference Matthews and Robbins2003; Mehta et al., Reference Mehta, Gore-Langton, Golembo, Colvert, Williams and Sonuga-Barke2010; Pechtel & Pizzagalli, Reference Pechtel and Pizzagalli2011; Phillips, Howes, Whitelaw, Robbins, & Everitt, Reference Phillips, Howes, Whitelaw, Robbins and Everitt1994; Pryce, Dettling, Spengler, Spaete, & Feldon, Reference Pryce, Dettling, Spengler, Spaete and Feldon2004). Individuals exposed to ELA are at a higher risk of substance and alcohol abuse disorders (Anda et al., Reference Anda, Croft, Felitti, Nordenberg, Giles, Williamson and Giovino1999; Dube et al., Reference Dube, Felitti, Dong, Chapman, Giles and Anda2003), which contribute to increased inflammation (Shiels et al., Reference Shiels, Katki, Freedman, Purdue, Wentzensen, Trabert and Chaturvedi2014). Though less relevant for children, adolescence might be a salient time for proinflammatory phenotypes to emerge due to increases in risky behaviors, such as smoking and habits that contribute to weight gain (Raposa, Bower, Hammen, Najman, & Brennan, Reference Raposa, Bower, Hammen, Najman and Brennan2014), and impulsive behaviors that increase stress exposure (Lovallo, Reference Lovallo2013; Raposa et al., Reference Raposa, Bower, Hammen, Najman and Brennan2014).

Another behavioral pathway that could follow from ELA to promote inflammation is through persistent or recurring distress and self-harming behaviors. Maltreatment has been associated with heightened threat perception (Leppanen & Nelson, Reference Leppanen and Nelson2009; Pollak & Kistler, Reference Pollak and Kistler2002) and heightened amygdala activation in response to negative emotional stimuli (McCrory et al., Reference McCrory, De Brito, Kelly, Bird, Sebastian, Mechelli and Viding2013; Tottenham et al., Reference Tottenham, Hare, Millner, Gilhooly, Zevin and Casey2011). Depression and inflammation are bidirectionally associated in longitudinal studies of adults and adolescents (Matthews et al., Reference Matthews, Schott, Bromberger, Cyranowski, Everson-Rose and Sowers2010; Miller & Cole, Reference Miller and Cole2012). An individual's perception of stressors, emotional symptoms, and behavioral responses when distressed could increase their inflammation levels (Danese & Lewis, Reference Danese and Lewis2017). An individual's increased exposure to stressors and their emotional symptoms can further lead to chronic levels of stress and individuals may engage in self-harm to relieve feelings of distress (Baldwin, Arseneault, Caspi et al., Reference Baldwin, Arseneault, Caspi, Moffitt, Fisher, Odgers and Danese2019), offering another behavioral pathway by which ELS can contribute to increased inflammation.

Sleep

There are reciprocal links between sleep and inflammation, and we direct the reader to a thorough review by Irwin (Reference Irwin2015). Sleep can regulate the immune response: sleep disturbance has been linked with reduced vaccine efficacy (e.g. Prather et al., Reference Prather, Hall, Fury, Ross, Muldoon, Cohen and Marsland2012; Spiegel, Sheridan, & Van Cauter, Reference Spiegel, Sheridan and Van Cauter2002), impaired response to infectious challenge (Cohen, Doyle, Alper, Janicki-Deverts, & Turner, Reference Cohen, Doyle, Alper, Janicki-Deverts and Turner2009; Toth, Reference Toth1995), and upregulation of inflammation (Irwin, Reference Irwin2015). Sleep influences the HPA axis, which in turn regulates the immune system through the diurnal cortisol rhythm (Irwin & Cole, Reference Irwin and Cole2011; Slavich & Irwin, Reference Slavich and Irwin2014). In experimental and naturalistic epidemiological studies, sleep deprivation is associated with increased expression of proinflammatory cytokines and elevated inflammation (Irwin, Wang, Campomayor, Collado-Hidalgo, & Cole, Reference Irwin, Wang, Campomayor, Collado-Hidalgo and Cole2006; Miller et al., Reference Miller, Kandala, Kivimaki, Kumari, Brunner, Lowe and Cappuccio2009). Maternal separation in rodent models decreased sleep duration and disrupted sleep architecture for stress-affected animals (Mrdalj et al., Reference Mrdalj, Pallesen, Milde, Jellestad, Murison, Ursin and Gronli2013). A history of childhood trauma is associated with increased sleep problems (Gregory & Sadeh, Reference Gregory and Sadeh2016), independent of current PTSD or depression diagnoses (Noll, Trickett, Susman, & Putnam, Reference Noll, Trickett, Susman and Putnam2006). Unfortunately, prospective evidence that demonstrates a mediating role for inflammation in the association between sleep disruptions and psychopathology is not yet available (Irwin, Reference Irwin2015). To our knowledge, no studies have extended this work to investigate the specific pathways between ELA and sleep disruption as they relate to inflammation and psychopathology.

Moderation

Meta-analyses of pediatric and adult populations have found heterogeneity in outcomes in the association of ELA and inflammation (Baumeister et al., Reference Baumeister, Akhtar, Ciufolini, Pariante and Mondelli2016; Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020). We propose three moderating factors that are currently underexamined in the literature: (a) age at inflammation assessment, (b) sex differences, and (c) population context differences. First, when are ELA's effects on inflammation instantiated and measurable? Do inflammatory phenotypes from ELA differ based on the timing of immune assessment? Second, there are questions on the timing of adversity exposure: are there sensitive periods for exposure to adversity and maximal impact on the immune system, or is a cumulative model of adversity exposure most salient to impacts on the immune system? Further, animals models of early life stress and inflammation routinely find sex differences (Ganguly & Brenhouse, Reference Ganguly and Brenhouse2015), though few studies examine sex differences in human populations. Finally, though much of the ELA and inflammation literature has been conducted with participants from Western, educated, industrialized, rich and democratic (“WEIRD”) societies, a handful of studies in different country contexts paint a different picture of the associations between ELA and inflammation. Thus, does societal context act as a yet-unexplored moderator? While studies and meta-analyses have tested for moderation in adult samples, more work is needed to understanding how moderation functions in pediatric samples.

Age of assessment

More research is needed to understand how ELA across development contributes to an inflammatory phenotype and how inflammatory phenotypes change across development. As noted, acute psychosocial stress activates peripheral inflammation and chronic psychosocial stress is associated with systemic, low-grade inflammation. This transition is thought to be a driver of psychopathology, especially after exposure to childhood adversity. However, it is unclear when an acute stressor becomes a chronic stressor (Rohleder, Nater, Wolf, Ehlert, & Kirschbaum, Reference Rohleder, Nater, Wolf, Ehlert and Kirschbaum2004), and this gap is especially prevalent in studies of children. Longitudinal assessments of inflammation over longer periods of time, repeat assessments of inflammation over a shorter window (for an example, see McDade et al., Reference McDade, Tallman, Madimenos, Liebert, Cepon, Sugiyama and Snodgrass2012), and studies on repeated acute stress might help fill this gap. As these studies do not yet exist, we review the ELA and inflammation literature with a developmental lens.

Adulthood

Much of the evidence linking ELA and later inflammation has been found in adults. Consistent with the animal literature, adults exposed to childhood maltreatment are at greater risk of increased levels of circulating inflammatory markers (Baumeister et al., Reference Baumeister, Akhtar, Ciufolini, Pariante and Mondelli2016; Danese & Lewis, Reference Danese and Lewis2017) and greater proinflammatory responses in response to subsequent stressors (Carpenter et al., Reference Carpenter, Gawuga, Tyrka, Lee, Anderson and Price2010; Gouin et al., Reference Gouin, Glaser, Malarkey, Beversdorf and Kiecolt-Glaser2012; Kiecolt-Glaser et al., Reference Kiecolt-Glaser, Gouin, Weng, Malarkey, Beversdorf and Glaser2011). In clinical adult populations, depressed individuals with a history of ELA exhibit increased inflammatory responses to acute psychosocial stress (Pace et al., Reference Pace, Mletzko, Alagbe, Musselman, Nemeroff, Miller and Heim2006) and show higher levels of inflammation in unstimulated plasma (Danese et al., Reference Danese, Moffitt, Pariante, Ambler, Poulton and Caspi2008). Table 2 collates the studies of adult clinical populations from the Baumeister meta-analysis, specifically focusing on populations with psychiatric diagnoses of first episode of psychosis (FEP), major depressive disorder (MDD), and PTSD as compared with health controls (Baumeister et al., Reference Baumeister, Akhtar, Ciufolini, Pariante and Mondelli2016). A history of ELA appears drive increased inflammation in clinical samples (Baumeister et al., Reference Baumeister, Akhtar, Ciufolini, Pariante and Mondelli2016), as patients without ELA had immune profiles similar to healthy controls (Dennison et al., Reference Dennison, McKernan, Cryan and Dinan2012). Further, psychologically healthy controls with a history of ELA exhibited higher IL-1B compared to controls without ELA exposure (Di Nicola et al., Reference Di Nicola, Cattaneo, Hepgul, Di Forti, Aitchison, Janiri and Mondelli2013).

Table 2. Early adversity and inflammation: adult at inflammation assessment, clinical populations

∅ = null association between ELA exposure and inflammatory outcome.

✓ = positive association between ELA exposure and inflammatory outcome.

CTQ = Child Trauma Questionnaire; CRP = C-reactive protein; ELA = early life adversity; FEP = first episode of psychosis; HC = “heathy controls”; IL-6 = interleukin-6; MDD = major depressive disorder; PTSD = posttraumatic stress disorder

The Baumeister meta-analysis found that age did not have a moderating effect on the relationship between ELA and inflammation in adulthood (Baumeister et al., Reference Baumeister, Akhtar, Ciufolini, Pariante and Mondelli2016). It could be possible that the moderating effect of age is most salient in pediatric populations. Alternatively, adulthood could be a life stage in which the cumulative effects of ELA are easier to capture through peripheral inflammatory cytokines. One study of adults in midlife found support for the stress accumulation model: ELA and recent life event stressors were independently associated with higher levels of inflammation, controlling for each other's effects, and their interaction was not significant (Hostinar, Lachman, Mroczek, Seeman, & Miller, Reference Hostinar, Lachman, Mroczek, Seeman and Miller2015a). In that study, mediation analyses found that ELA and recent life stressors had unique and additive contributions to inflammation through norepinephrine pathways, adiposity, and inflammation-inducing behaviors (Hostinar et al., Reference Hostinar, Lachman, Mroczek, Seeman and Miller2015a). In adults, inflammation might depend on behavioral mediators rather than age.

Childhood and adolescence

Descriptive analyses from the meta-analysis of ELA and inflammation associations in a pediatric population found that adversity was positively associated with CRP when inflammation was assessed in adolescence, but not with IL-6 or TNF-α (Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020). A study of 1,789 female adolescents found that greater exposure to childhood maltreatment and victimization predicted higher CRP levels at age 18 (Baldwin et al., Reference Baldwin, Arseneault, Caspi, Fisher, Moffitt, Odgers and Danese2018). This study included prospectively collected information from 5 to 12 years of age about ELA exposure, but the study took a cumulative approach rather than testing ELA exposure at different developmental periods. A study of sexual abuse in a clinic setting found that individuals getting a medical assessment for sexual abuse (occurring in the 72 hr preceding the assessment) did exhibit higher levels of IL-6 than their nonabused peers (Kilic et al., Reference Kilic, Ozlem and Murat2017). However, the sample size was small and, while the mean age of assessment was 15 years, the age range spanned from 3 to 18 years, which makes it difficult to interpret as it pertains to time in development. Given the temporal proximity of the sexual abuse to the inflammation metric, it is unclear if this study is indicative of acute physical injury or chronic inflammation.

The pediatric meta-analysis previously mentioned also found in descriptive analyses that ELA was positively associated with CRP when inflammation was assessed in later childhood (Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020). Two found positive associations with maltreatment exposure and circulating levels of TNF-α, but not IL-6 (Bucker et al., Reference Bucker, Fries, Kapczinski, Post, Yatham, Vianna and Kauer-Sant'Anna2015). In contrast, a larger study of 9-year-olds exposed to maltreatment found no associations between ELA and salivary CRP, even when examining the contributions of timing and chronicity of exposure (Cicchetti et al., Reference Cicchetti, Handley and Rogosch2015). It is difficult to compare the studies, as the inflammatory cytokines and tissue selection differed. However, both studies measured maltreatment prospectively and measured inflammation at age 9, highlighting the need for more studies on ELA and inflammation in childhood.

Few studies have examined ELA and inflammation in infancy and early childhood but descriptive analyses from the previously mentioned meta-analyses found that ELA was positively associated with CRP in infancy but negatively associated with CRP in early childhood (Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020). This meta-analysis included studies with a very broad definition of ELA that are not included in our definition of victimization. A study of salivary CRP in 40 4-year-olds found no association between ELA and CRP, though ELA was positively associated with salivary IL-1β (Tyrka et al., Reference Tyrka, Parade, Valentine, Eslinger and Seifer2015). In a study using dried blood spot measures of CRP, no group differences were found in CRP levels between 4–6 year old CPS-referred children and low-risk children (Bernard et al., Reference Bernard, Hostinar and Dozier2019). The study did find that both insecure and disorganized attachments in CPS-referred children were associated with more inflammation compared to securely attached CPS-referred children and low-risk children (Bernard et al., Reference Bernard, Hostinar and Dozier2019).

The Avon Longitudinal Study of Parents and Children (ALSPAC) study, a prospective cohort study of 4,525 individuals with data on IL-6 and CRP in childhood (age 9 years), provides a wealth of longitudinal data on ELA, inflammation, and psychopathology-related outcomes. One analysis found that prospective measures of adversity predicted IL-6 but not CRP in 9-year-olds (Flouri et al., Reference Flouri, Francesconi, Midouhas and Lewis2020). IL-6 partially mediated the relationship between ELA and later internalizing symptoms (Flouri et al., Reference Flouri, Francesconi, Papachristou, Midouhas and Lewis2019). However, IL-6 did not explain the association between ELA and externalizing symptoms. A separate study on this cohort found that IL-6 levels at 9 years partially mediated the association between ELA in childhood and depressive symptoms at the beginning of adolescence (Flouri et al., Reference Flouri, Francesconi, Papachristou, Midouhas and Lewis2019). Internalizing symptoms at age 9 years fully mediated the association between levels of IL-6 in childhood and subclinical psychotic symptoms (psychotic-like experiences, PLEs) at age 18 (Flouri et al., Reference Flouri, Francesconi, Midouhas and Lewis2020). CRP was not associated with either internalizing symptoms or psychotic-like experiences. Though it is unclear how IL-6 tracks across development, these studies provide evidence that that specific markers of inflammation in childhood mediate the relationship between ELA and symptoms of psychopathology.

Timing and duration of exposure

Cumulative adversity throughout childhood may be needed to see associations with inflammation. Early intervention and removal from adversity may protect an individual from developing an inflammatory phenotype and instead instantiate risk for psychopathology through other mechanisms. As mentioned previously, studies of adolescents and young adults removed institutional care indicate no evidence of a proinflammatory phenotype after exposure to deprivation (Reid et al., Reference Reid, Coe, Doyle, Sheerar, Slukvina, Donzella and Gunnar2019; Slopen et al., Reference Slopen, Tang, Nelson, Zeanah, McDade, McLaughlin and Fox2019; Tang et al., Reference Tang, Slopen, Nelson, Zeanah, Georgieff and Fox2018). As these studies did find changes in acquired immunity indicative of immune senescence (Elwenspoek et al., Reference Elwenspoek, Hengesch, Leenen, Schritz, Sias, Schaan and Muller2017a; Reference Elwenspoek, Sias, Hengesch, Schaan, Leenen, Adams and Turner2017b; Reid et al., Reference Reid, Coe, Doyle, Sheerar, Slukvina, Donzella and Gunnar2019), adolescence might be a time to capture early perturbations to acquired immunity that are either on the path to or distinct from the pathogenesis of inflammation and psychopathology. In the Avon Longitudinal Study of Parents and Children study mentioned previously, there was also no evidence that IL-6 or CRP levels mediated the association between ELA exposure in infancy and depressive symptoms in adolescence (Flouri et al., Reference Flouri, Francesconi, Midouhas and Lewis2020). The link from ELA in infancy to depressive symptoms in adolescence was direct, and therefore elevated levels of IL-6 in childhood partially explained the link between adolescent depressive symptoms from proximal childhood stressors only (Flouri et al., Reference Flouri, Francesconi, Midouhas and Lewis2020). Thus, there may be qualitatively different pathways to depression from very early ELA exposure. Associations between ELA and inflammation may not emerge without substantial environmental or behavioral input (Miller et al., Reference Miller, Chen and Parker2011b). Future studies that address the auto-correlation of early and concurrent adversity can help tease apart how the timing of childhood adversity contributes to increased risk of psychopathology and inflammation.

Summary

What does it mean, functionally and regarding psychopathology risk, to have higher levels of inflammatory cytokines in infancy versus childhood versus adolescence? Relatively little is known about the relationship between adversity and inflammation longitudinally from infancy to adolescence. Even less is known about the pathogenesis of inflammation from ELA in infants and young children. Thus, given the meta-analytical findings that show increased inflammation in infancy, no association in childhood, and increased inflammation in adolescence, it is unclear whether the association between ELA and inflammation across development is linear or nonlinear. This might suggest support for theoretical sensitive periods of immune development in infants and adolescents (Kuhlman et al., Reference Kuhlman, Horn, Chiang and Bower2020). However, the picture becomes less clear when examining ELA exposure limited to infancy compared to cumulative exposure. Developmentally focused research is needed to understand the “hidden wounds” of ELA.

Sex differences

Experimental, nonhuman animal models suggest that early life stress has sexually dimorphic effects on immune dysregulation (Ganguly & Brenhouse, Reference Ganguly and Brenhouse2015). In adult populations, a meta-analysis found no evidence of a moderating effect of sex on the relationship between ELA and inflammation (Baumeister et al., Reference Baumeister, Akhtar, Ciufolini, Pariante and Mondelli2016). The pediatric meta-analysis was underpowered to examine sex differences. Nonetheless, the association between ELA and CRP was specific only to young women in the genetically informed twin study mentioned previously (Baldwin et al., Reference Baldwin, Arseneault, Caspi, Fisher, Moffitt, Odgers and Danese2018). Sex differences could arise from sex differences in neuroendocrine regulation. For example, male, not female, adolescents exposed to violence in the past year exhibited lower cortisol reactivity to a laboratory stressor (Peckins, Dockray, Eckenrode, Heaton, & Susman, Reference Peckins, Dockray, Eckenrode, Heaton and Susman2012). Sleep disruption may be another avenue by which sex moderates the relationship between ELA, inflammation, and psychopathology. Women appear more likely to develop dysregulated inflammatory responses from sleep disturbance while men appear to be more likely to develop cardiovascular disease and cancer, but not depression (Irwin, Reference Irwin2015). The field needs studies that are well-powered to examine sex differences in pediatric populations, as exposure to ELA and the biological sequalae of inflammation could exhibit important sexually dimorphic effects in humans.

Ecological and demographic differences

Examination of heterogeneous populations and contexts could reveal important mechanisms of how ELA contributes to inflammation and psychopathology. Many of the studies reviewed have been conducted in high-income country contexts, though children in low- and middle-income country contexts are no less exposed to ELA. However, the processes by which ELA may instantiate inflammation might be different for children outside of high-income country and often urban contexts. Studies in representative cohorts in the Philippines, as mentioned previously, and Tanzania suggest that the typical high-adversity, high-inflammation associations may be moderated by contextual factors that are unique to high-microbial, rural, or low- or middle-income country contexts (Hadley & Decaro, Reference Hadley and Decaro2014; McDade, Hoke, Borja, Adair, & Kuzawa, Reference McDade, Hoke, Borja, Adair and Kuzawa2013). In a pediatric population, a large study of Tanzanian children under 5 found that CRP was positively associated with adversity quantified as maternal illiteracy, larger household size, and disease load (Hadley & Decaro, Reference Hadley and Decaro2014). Interestingly, membership in the least poor wealth category was associated with higher levels of CRP. As this sample of Tanzanian children were more likely to be stunted, with an average height-for-age of −1.66, and an average CRP level of 4.4 mg/l, it is difficult to compare with the studies of infants conducted in higher-income countries (Hadley & Decaro, Reference Hadley and Decaro2014). However, this study points to important contextual considerations when researching young children, ELA, and inflammatory biomarkers in more diverse country contexts.

Even in the context of Western, educated, industrialized, rich and democratic (WEIRD) societies, under-represented ethnic and race groups in predominantly white countries might experience differences in context that contribute to increases in inflammation after ELA exposure. For example, Pereira's analysis of the large US cohort mentioned previously found that sexual abuse was associated with all inflammatory markers in nonwhite adults but not white adults in the United States. IL-6 was higher by 36.3% in nonwhite adults exposed to childhood sexual abuse versus 9.89% in white adults exposed to childhood sexual abuse (Pinto Pereira et al., Reference Pinto Pereira, Stein Merkin, Seeman and Power2019). Experiences of racial or ethnic discrimination may constitute an aspect of child victimization or threat perception that is currently understudied. In a study of perceived discrimination, there was a positive association between child-reported discrimination and CRP, as measured by dried blood spots in a group of children with a mean age of 12 y (Goosby, Malone, Richardson, Cheadle, & Williams, Reference Goosby, Malone, Richardson, Cheadle and Williams2015). This was a small study, but emphasizes the need to operationalize, measure, and assess how different forms of child adversity, such as racial discrimination, can contribute to inflammatory outcomes and risk for psychopathology. Thus, specific populations may experience moderating factors such as discrimination in addition to the types of adversity that are typically measured in the context of childhood adversity exposure and inflammation (Goosby et al., Reference Goosby, Malone, Richardson, Cheadle and Williams2015; Pinto Pereira et al., Reference Pinto Pereira, Stein Merkin, Seeman and Power2019; Slopen et al., Reference Slopen, Lewis, Gruenewald, Mujahid, Ryff, Albert and Williams2010).

Interventions

The accumulated evidence suggests that the study of inflammation could help explain and plan a course of treatment for individuals with psychopathology after exposure to ELA. Nevertheless, there are a number of questions that can only be answered with animal models. For example, animal models could help elucidate developmental pathways from ELA and inflammation, assist in biomarker selection, clarify tissue specificity issues. As ELA is typically associated with later adversity in humans, animal models can help answer questions about the timing of inflammation assessment. In humans, clinical interventions can begin to address the heterogeneity in outcomes that might be experience- or population-dependent. Lifestyle and psychosocial interventions can begin to clarify the outstanding questions on mediation. Here, we address some of the evidence from human intervention models.

Clinical interventions

There is still much to understand about treating inflammation-associated psychopathologies with anti-inflammatory medications and supplements. Experimental studies on anti-inflammatory treatments in depression offer promise, especially in subgroups of patients who exhibit high levels of inflammation in the case in ELA-exposed individuals with depression (Danese & Lewis, Reference Danese and Lewis2017; McIntyre et al., Reference McIntyre, Subramaniapillai, Lee, Pan, Carmona, Shekotikhina and Mansur2019; Raison et al., Reference Raison, Rutherford, Woolwine, Shuo, Schettler, Drake and Miller2013). Meta-analyses examining inflammation before and after treatment in depressed patients found that IL-6 levels decreased with antidepressant treatment regardless of outcome, whereas elevated TNFα was associated with treatment resistance (Strawbridge et al., Reference Strawbridge, Arnone, Danese, Papadopoulos, Herane Vives and Cleare2015). Patients who were nonresponders had higher baseline inflammation, suggesting that elevated levels of inflammation contribute to treatment resistance (Strawbridge et al., Reference Strawbridge, Arnone, Danese, Papadopoulos, Herane Vives and Cleare2015). Studies of major depressive disorder and bipolar disorder patients with high levels of systemic inflammation and a history of physical abuse demonstrated a superior response to the TNF-α blocker infliximab (McIntyre et al., Reference McIntyre, Subramaniapillai, Lee, Pan, Carmona, Shekotikhina and Mansur2019). This initial evidence points to the potential benefit of pharmacological anti-inflammatory interventions in patients’ subgroups but requires further investigation before any clinical implementation.

Lifestyle interventions

Beyond clinical interventions, diverse lifestyle interventions could reduce inflammation and either prevent or reduce psychopathology symptoms. Ideally, lifestyle interventions such as diet, physical activity, and sleep can be tailored and used across development with minimal adverse side effects.

In the last decade, numerous studies report inverse associations between diet quality and depressive disorders and symptoms in adults (Jacka, Mykletun, Berk, Bjelland, & Tell, Reference Jacka, Mykletun, Berk, Bjelland and Tell2011b; Jacka et al., Reference Jacka, Pasco, Williams, Meyer, Digger and Berk2013; Kuczmarski et al., Reference Kuczmarski, Cremer Sees, Hotchkiss, Cotugna, Evans and Zonderman2010; Nanri et al., Reference Nanri, Kimura, Matsushita, Ohta, Sato, Mishima and Mizoue2010), adolescents (Jacka et al., Reference Jacka, Kremer, Berk, de Silva-Sanigorski, Moodie, Leslie and Swinburn2011a; Jacka et al., Reference Jacka, Kremer, Leslie, Berk, Patton, Toumbourou and Williams2009; Weng et al., Reference Weng, Hao, Qian, Cao, Fu, Sun and Tao2012), and children (Kohlboeck et al., Reference Kohlboeck, Sausenthaler, Standl, Koletzko, Bauer and von Berg2012). Diet quality is associated with inflammation in population-based studies (Chrysohoou, Panagiotakos, Pitsavos, Das, & Stefanadis, Reference Chrysohoou, Panagiotakos, Pitsavos, Das and Stefanadis2004; Fung et al., Reference Fung, Rimm, Spiegelman, Rifai, Tofler, Willett and Hu2001; Lopez-Garcia et al., Reference Lopez-Garcia, Schulze, Fung, Meigs, Rifai, Manson and Hu2004) and intervention studies (Esposito et al., Reference Esposito, Marfella, Ciotola, Di Palo, Giugliano, Giugliano and Giugliano2004; Watzl, Kulling, Moseneder, Barth, & Bub, Reference Watzl, Kulling, Moseneder, Barth and Bub2005). These associations may be due to specific aspects of diet quality that either contribute to or protect from inflammatory processes.

For example, magnesium intake is highly correlated with diet quality and is negatively associated with levels of CRP (King, Mainous, Geesey, & Woolson, Reference King, Mainous, Geesey and Woolson2005). Omega-3 fatty acids and polyunsaturated fatty acids are associated with reduced inflammation (Clarke, Shipley, Armitage, Collins, & Harris, Reference Clarke, Shipley, Armitage, Collins and Harris2009; Rangel-Huerta, Aguilera, Mesa, & Gil, Reference Rangel-Huerta, Aguilera, Mesa and Gil2012), and a randomized controlled trial of omega-3 fatty acids found a reduction in psychotic disorder progression and psychiatric morbidity over a 7-year follow-up (Amminger, Schafer, Schlogelhofer, Klier, & McGorry, Reference Amminger, Schafer, Schlogelhofer, Klier and McGorry2015). Foods high in fiber could have positive impacts on immune functioning due to high levels of beta-glucans, phytochemicals, and influences on the gut microbiome (Bilici et al., Reference Bilici, Efe, Koroglu, Uydu, Bekaroglu and Deger2001; Parikh et al., Reference Parikh, Pollock, Bhagatwala, Guo, Gutin, Zhu and Dong2012; Tachon, Zhou, Keenan, Martin, & Marco, Reference Tachon, Zhou, Keenan, Martin and Marco2013; Volman, Ramakers, & Plat, Reference Volman, Ramakers and Plat2008). As noted in the nutrient metabolism mechanism section, adequate iron intake during infancy may act as a protective factor from increased levels of CRP in adolescence (Reid et al., Reference Reid, Doom, Argote, Correa-Burrows, Lozoff, Blanco and Gahagan2020). In contrast, diets high in refined carbohydrates, added sugars, and trans-fatty acids are associated with higher levels of inflammation in human and animal models (Iwata et al., Reference Iwata, Pham, Rizzo, Cheng, Maloney and Kim2011; Levitan et al., Reference Levitan, Cook, Stampfer, Ridker, Rexrode, Buring and Liu2008; Liu et al., Reference Liu, Manson, Buring, Stampfer, Willett and Ridker2002; Pistell et al., Reference Pistell, Morrison, Gupta, Knight, Keller, Ingram and Bruce-Keller2010). Saturated and trans-fat intake may increase LPS release from microbiota in the gut to promote immune system activation (Moreira, Texeira, Ferreira, Peluzio Mdo, & Alfenas Rde, Reference Moreira, Texeira, Ferreira, Peluzio Mdo and Alfenas Rde2012). More studies on adversity, diet, and inflammation are needed in adult and child cohorts to test the impact of dietary interventions in ELA-exposed populations.

Physical activity is another area of intervention. Exercise can reduce depressive symptoms (Conn, Reference Conn2010) and the risk of new depressive episodes in adults (Brown, Ford, Burton, Marshall, & Dobson, Reference Brown, Ford, Burton, Marshall and Dobson2005; Pasco et al., Reference Pasco, Williams, Jacka, Henry, Coulson, Brennan and Berk2011; Strawbridge, Deleger, Roberts, & Kaplan, Reference Strawbridge, Deleger, Roberts and Kaplan2002). Physical inactivity in childhood has been associated with an increased risk of adult depression (Jacka et al., Reference Jacka, Pasco, Williams, Leslie, Dodd, Nicholson and Berk2011c). A longitudinal study of 3,809 nondepressed men and women found that physical activity mediated the association between inflammation and depressive symptoms (Frank et al., Reference Frank, Kaushal, Poole, Lawes, Chalder and Cadar2019). A meta-analysis of 83 studies found that exercise training was negatively associated with CRP levels regardless of the age or sex of the individual (Fedewa, Hathaway, & Ward-Ritacco, Reference Fedewa, Hathaway and Ward-Ritacco2017). Few studies have been conducted in children, and most studies are in overweight or obese samples of children (Michigan, Johnson, & Master, Reference Michigan, Johnson and Master2011). Physical activity is a modifiable health behavior that could be used to target inflammation and depressive symptoms. However, to our knowledge, no studies have investigated physical activity and inflammation in the context of ELA.

Behavioral interventions that target sleep have the potential to reduce inflammation and possibly reduce inflammatory disease risk in contexts of psychopathology (Irwin, Reference Irwin2015). Sleep health can theoretically be used to therapeutically control chronic infectious, inflammatory, and neuropsychiatric diseases (Irwin, Reference Irwin2015). Several behavioral (e.g., cognitive behavioral therapy for insomnia) and mind–body (e.g., tai chi and yoga) interventions have potential to modulate certain aspects of the immune functioning. For example, mind–body interventions have been found to improve insomnia (Irwin, Olmstead, & Motivala, Reference Irwin, Olmstead and Motivala2008), augment vaccine responses (Irwin, Olmstead, & Oxman, Reference Irwin, Olmstead and Oxman2007), and reduce levels of IL-6 (Irwin & Olmstead, Reference Irwin and Olmstead2012), and reduce proinflammatory response gene profiles (Creswell et al., Reference Creswell, Irwin, Burklund, Lieberman, Arevalo, Ma and Cole2012). Similarly, behavioral therapy in patients with rheumatoid arthritis improved depressive symptoms, reduced the stimulated production of the proinflammatory cytokine IL-6 (Zautra et al., Reference Zautra, Davis, Reich, Nicassario, Tennen, Finan and Irwin2008), and decreased levels of CRP up to a year after treatment (Irwin et al., Reference Irwin, Olmstead, Carrillo, Sadeghi, Breen, Witarama and Nicassio2014). Interventions might be specifically helpful for adolescents, as a study of 315 adolescents found that nightly variability in sleep duration was associated with higher levels of CRP (Park et al., Reference Park, Tsai, Dahl, Irwin, McCreath, Seeman and Fuligni2016). For younger adolescents, shorter average sleep duration was associated with higher CRP (Park et al., Reference Park, Tsai, Dahl, Irwin, McCreath, Seeman and Fuligni2016). While more studies are needed in younger children and infants, this suggests that targeting sleep duration could reduce inflammation following ELA.

Psychosocial interventions

Interventions to reduce the incidence of ELA itself are clearly necessary to reduce the rates of mental and physical illness. However, interventions in the context of ongoing childhood adversity show promise in reducing harmful outcomes. A study of positive childhood experiences in a cross-sectional study of 6,188 adults found that self-reports of positive childhood experiences showed a dose–response association with lower odds of adult depression and/or poor mental health (Bethell, Jones, Gombojav, Linkenbach, & Sege, Reference Bethell, Jones, Gombojav, Linkenbach and Sege2019). These associations held after controlling for exposure to adverse childhood experiences, and support the prioritization of providing children with safe, stable nurturing relationships for better mental health outcomes (Bethell et al., Reference Bethell, Jones, Gombojav, Linkenbach and Sege2019). Positive family and caregiving environments are associated with low inflammation across a range of developmental ages. A study in infants found that attachment security moderated the association between ELA and inflammation, where securely attached infants did not exhibit a heightened inflammatory phenotype (Measelle & Ablow, Reference Measelle and Ablow2018). As noted earlier, attachment quality in infancy has been found to predict early childhood CRP among CPS referred children (Bernard et al., Reference Bernard, Hostinar and Dozier2019). Children who had insecure or disorganized attachments in infancy had higher CRP levels in early childhood than children who had secure attachments in infancy, controlling for covariates (Bernard et al., Reference Bernard, Hostinar and Dozier2019). Interventions such as the Attachment and Biobehavioral Catch-up (ABC) intervention have demonstrated efficacy in improving attachment quality in families at high risk of developing insecure attachment (Bernard et al., Reference Bernard, Dozier, Bick, Lewis-Morrarty, Lindhiem and Carlson2012), and parenting interventions early in life therefore may be a promising avenue to pursue reducing later inflammatory phenotypes.

In addition to prevention, the therapeutic improvement of social support and community belonging could be promising in reducing the associations between child adversity and adult inflammatory outcomes. One study investigated whether belonging to the tribal community in a sample of 90 adults living on the Blackfeet reservation may moderate the relationship between ELA and levels of IL-6 and CRP (John-Henderson et al., Reference John-Henderson, Henderson-Matthews, Ollinger, Racine, Gordon, Higgins and Rynda-Apple2020). Independent of age, gender, annual income, body mass index, and depressive symptoms, ELA exposure predicted IL-6 and CRP levels, but self-reports of belonging to the tribal community moderated the relationship between reports of ELA and markers of inflammation (John-Henderson et al., Reference John-Henderson, Henderson-Matthews, Ollinger, Racine, Gordon, Higgins and Rynda-Apple2020). Outside of parenting interventions, one study found that the presence of supportive role models and positive cognitive models to deal with stress may act as a buffer against proinflammatory phenotypes for adolescents experiencing adversity (Chen, Lee, Cavey, & Ho, Reference Chen, Lee, Cavey and Ho2013). In this study, youth with supportive role models had lower IL-6 levels. Youth who were better able to reframe stressors a benign and better able to persisting with optimism (“shift-and-persist”) had lower IL-6 (Chen et al., Reference Chen, Lee, Cavey and Ho2013). These benefits seemed to especially benefit the low-SES youth, as benefits were not found among high-SES adolescents (Chen et al., Reference Chen, Lee, Cavey and Ho2013). This study suggests that identifying psychological buffers in adolescents has could have implications for inflammation levels and possibly mental health disparities. Despite many possible avenues of intervention and prevention, more research is needed in this space and in the basic science of understanding how childhood adversity contributes to psychopathology through inflammatory pathways.

Conclusion

Exposure to ELA is a critical risk factor for the development of psychopathology, and the understanding of how early adversity gets “under the skin” is growing with increased research on the biological risk for increased inflammation. Nevertheless, the heterogeneity present in the current literature leaves many questions and many opportunities for future research. Studies that are developmentally informed and grounded in an understanding of immune- and stress-mediating systems are needed as the field grows. In addition to population-based studies, mechanistic and experimental work is needed to understand what increased inflammation means in the context of adversity at specific times in development and longitudinally. In addition to a better understanding of moderation and measurement, more research is needed to elucidate how ELA contributes to inflammation through neuroendocrine dysregulation, brain functioning and behavior, sleep, adipose tissue, the gut microbiome, and stress-induced nutrient dysregulation. All of these possible mechanistic pathways emphasize the myriad intervention and prevention routes that could be pursued to reduce the mental health burden on individuals and communities that arises from the hidden wounds of childhood adversity.

Acknowledgments

The views expressed are those of the authors and not necessarily those of the NSF, the NHS, the NIHR, the Department of Health and Social Care.

Funding Statement

BR was funded by the National Science Foundation (NSF; grant 00039202) and the University of Minnesota Doctoral Dissertation Fellowship. AD was funded by the Medical Research Council (MRC; grant no. P005918) and the National Institute for Health Research (NIHR) Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London.

Conflicts of Interest

None.

References

Agnew-Blais, J., & Danese, A. (2016). Childhood maltreatment and unfavourable clinical outcomes in bipolar disorder: A systematic review and meta-analysis. Lancet Psychiatry, 3, 342349. doi:10.1016/S2215-0366(15)00544-1CrossRefGoogle ScholarPubMed
Amminger, G. P., Schafer, M. R., Schlogelhofer, M., Klier, C. M., & McGorry, P. D. (2015). Longer-term outcome in the prevention of psychotic disorders by the Vienna omega-3 study. Nature Communications, 6, 7934. doi:10.1038/ncomms8934CrossRefGoogle ScholarPubMed
Anda, R. F., Croft, J. B., Felitti, V. J., Nordenberg, D., Giles, W. H., Williamson, D. F., & Giovino, G. A. (1999). Adverse childhood experiences and smoking during adolescence and adulthood. JAMA, 282, 16521658. doi:10.1001/jama.282.17.1652CrossRefGoogle ScholarPubMed
Atladottir, H. O., Thorsen, P., Ostergaard, L., Schendel, D. E., Lemcke, S., Abdallah, M., & Parner, E. T. (2010). Maternal infection requiring hospitalization during pregnancy and autism spectrum disorders. Journal of Autism and Developmental Disorders, 40, 14231430. doi:10.1007/s10803-010-1006-yCrossRefGoogle ScholarPubMed
Avitsur, R., Hunzeker, J., & Sheridan, J. F. (2006). Role of early stress in the individual differences in host response to viral infection. Brain Behavior & Immunity, 20, 339348. doi:10.1016/j.bbi.2005.09.006CrossRefGoogle ScholarPubMed
Bailey, M. T., & Coe, C. L. (1999). Maternal separation disrupts the integrity of the intestinal microflora in infant rhesus monkeys. Developmental Psychobiology, 35, 146155. doi:10.1002/(sici)1098-2302(199909)35:2<146::aid-dev7>3.0.co;2-g3.0.CO;2-G>CrossRefGoogle ScholarPubMed
Baldwin, J. R., Arseneault, L., Caspi, A., Fisher, H. L., Moffitt, T. E., Odgers, C. L., … Danese, A. (2018). Childhood victimization and inflammation in young adulthood: A genetically sensitive cohort study. Brain Behavior & Immunity, 67, 211217. doi:10.1016/j.bbi.2017.08.025CrossRefGoogle ScholarPubMed
Baldwin, J. R., Arseneault, L., Caspi, A., Moffitt, T. E., Fisher, H. L., Odgers, C. L., … Danese, A. (2019). Adolescent victimization and self-injurious thoughts and behaviors: A genetically sensitive cohort study. Journal of the American Academy of Child and Adolescent Psychiatry, 58, 506513. doi:10.1016/j.jaac.2018.07.903CrossRefGoogle ScholarPubMed
Baldwin, J. R., & Danese, A. (2019). Pathways from childhood maltreatment to cardiometabolic disease: A research review. Adoption & Fostering, 43, 329339. doi:10.1177/0308575919856175CrossRefGoogle Scholar
Baldwin, J. R., Reuben, A., Newbury, J. B., & Danese, A. (2019). Agreement between prospective and retrospective measures of childhood maltreatment: A systematic review and meta-analysis. JAMA Psychiatry, 76, 584593. doi:10.101/jamapsychiatry.2019.0097CrossRefGoogle ScholarPubMed
Barnes, P. J., & Adcock, I. M. (2009). Glucocorticoid resistance in inflammatory diseases. Lancet, 373, 19051917. doi:10.1016/S0140-6736(09)60326-3CrossRefGoogle ScholarPubMed
Baumeister, D., Akhtar, R., Ciufolini, S., Pariante, C. M., & Mondelli, V. (2016). Childhood trauma and adulthood inflammation: A meta-analysis of peripheral C-reactive protein, interleukin-6 and tumour necrosis factor-alpha. Molecular Psychiatry, 21, 642649. doi:10.1038/mp.2015.67CrossRefGoogle ScholarPubMed
Beattie, E. C., Stellwagen, D., Morishita, W., Bresnahan, J. C., Ha, B. K., Von Zastrow, M., … Malenka, R. C. (2002). Control of synaptic strength by glial TNFalpha. Science, 295, 22822285. doi:10.1126/science.1067859CrossRefGoogle ScholarPubMed
Berk, M., Kapczinski, F., Andreazza, A. C., Dean, O. M., Giorlando, F., Maes, M., … Malhi, G. S. (2011). Pathways underlying neuroprogression in bipolar disorder: focus on inflammation, oxidative stress and neurotrophic factors. Neuroscience & Biobehavioral Reviews, 35, 804817. doi:10.1016/j.neubiorev.2010.10.001CrossRefGoogle ScholarPubMed
Bernard, K., Dozier, M., Bick, J., Lewis-Morrarty, E., Lindhiem, O., & Carlson, E. (2012). Enhancing attachment organization among maltreated children: Results of a randomized clinical trial. Child Development, 83, 623636. doi:10.1111/j.1467-8624.2011.01712.xCrossRefGoogle ScholarPubMed
Bernard, K., Hostinar, C. E., & Dozier, M. (2015). Intervention effects on diurnal cortisol rhythms of Child Protective Services-referred infants in early childhood: Preschool follow-up results of a randomized clinical trial. JAMA Pediatrics, 169, 112119. doi:10.1001/jamapediatrics.2014.2369CrossRefGoogle ScholarPubMed
Bernard, K., Hostinar, C. E., & Dozier, M. (2019). Longitudinal associations between attachment quality in infancy, C-reactive protein in early childhood, and BMI in middle childhood: Preliminary evidence from a CPS-referred sample. Attachment & Human Development, 21, 522. doi:10.1080/14616734.2018.1541513CrossRefGoogle ScholarPubMed
Bertone-Johnson, E. R., Whitcomb, B. W., Missmer, S. A., Karlson, E. W., & Rich-Edwards, J. W. (2012). Inflammation and early-life abuse in women. American Journal of Preventive Medicine, 43, 611620. doi:10.1016/j.amepre.2012.08.014CrossRefGoogle ScholarPubMed
Besedovsky, H., del Rey, A., Sorkin, E., & Dinarello, C. A. (1986). Immunoregulatory feedback between interleukin-1 and glucocorticoid hormones. Science, 233, 652654. doi:10.1126/science.3014662CrossRefGoogle ScholarPubMed
Bethell, C., Jones, J., Gombojav, N., Linkenbach, J., & Sege, R. (2019). Positive childhood experiences and adult mental and relational health in a statewide sample: Associations across adverse childhood experiences levels. JAMA Pediatrics, 173, e193007. doi:10.1001/jamapediatrics.2019.3007CrossRefGoogle Scholar
Bilbo, S. D., & Schwarz, J. M. (2009). Early-life programming of later-life brain and behavior: A critical role for the immune system. Frontiers in Behavioral Neuroscience, 3, 14. doi:10.3389/neuro.08.014.2009CrossRefGoogle ScholarPubMed
Bilici, M., Efe, H., Koroglu, M. A., Uydu, H. A., Bekaroglu, M., & Deger, O. (2001). Antioxidative enzyme activities and lipid peroxidation in major depression: Alterations by antidepressant treatments. Journal of Affective Disorders, 64, 4351. doi:10.1016/s0165-0327(00)00199-3CrossRefGoogle ScholarPubMed
Black, P. H. (2003). The inflammatory response is an integral part of the stress response: Implications for atherosclerosis, insulin resistance, type II diabetes and metabolic syndrome X. Brain, Behavior, and Immunity, 17, 350364. doi:10.1016/s0889-1591(03)00048-5CrossRefGoogle ScholarPubMed
Bosch, J. A. (2014). The use of saliva markers in psychobiology: Mechanisms and methods. Monographs in Oral Science, 24, 99108. doi:10.1159/000358864CrossRefGoogle ScholarPubMed
Boulanger, L. M. (2009). Immune proteins in brain development and synaptic plasticity. Neuron, 64, 93109. doi:10.1016/j.neuron.2009.09.001CrossRefGoogle ScholarPubMed
Brenhouse, H. C., & Thompson, V. (2015). Maternal separation increases IBA-1 expression: a microglia activation marker in the prefrontal cortex of adolescent males following a second hit of stress. Paper presented at the Society for Biological Psychiatry.Google Scholar
Brown, W. J., Ford, J. H., Burton, N. W., Marshall, A. L., & Dobson, A. J. (2005). Prospective study of physical activity and depressive symptoms in middle-aged women. American Journal of Preventative Medicine, 29, 265272. doi:10.1016/j.amepre.2005.06.009CrossRefGoogle ScholarPubMed
Bruce, J., Fisher, P. A., Pears, K. C., & Levine, S. (2009). Morning cortisol Levels in preschool-aged foster children: Differential effects of maltreatment type. Developmental Psychobiology, 51, 1423. doi:10.1002/dev.20333CrossRefGoogle ScholarPubMed
Brunson, K. L., Kramar, E., Lin, B., Chen, Y., Colgin, L. L., Yanagihara, T. K., … Baram, T. Z. (2005). Mechanisms of late-onset cognitive decline after early-life stress. Journal of Neuroscience, 25, 93289338. doi:10.1523/JNEUROSCI.2281-05.2005CrossRefGoogle ScholarPubMed
Bucker, J., Fries, G. R., Kapczinski, F., Post, R. M., Yatham, L. N., Vianna, P., … Kauer-Sant'Anna, M. (2015). Brain-derived neurotrophic factor and inflammatory markers in school-aged children with early trauma. Acta Psychiatrica Scandinavica, 131, 360368. doi:10.1111/acps.12358CrossRefGoogle ScholarPubMed
Bzostek, S. H., & Beck, A. N. (2011). Familial instability and young children's physical health. Social Science & Medicine, 73, 282292. doi:10.1016/j.socscimed.2011.04.014CrossRefGoogle Scholar
Callaghan, B. L., Fields, A., Gee, D. G., Gabard-Durnam, L., Caldera, C., Humphreys, K. L., … Tottenham, N. (2020). Mind and gut: Associations between mood and gastrointestinal distress in children exposed to adversity. Development and Psychopathology, 32, 309328. doi:10.1017/S0954579419000087CrossRefGoogle ScholarPubMed
Cani, P. D., Osto, M., Geurts, L., & Everard, A. (2012). Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity. Gut Microbes, 3, 279288. doi:10.4161/gmic.19625CrossRefGoogle ScholarPubMed
Capuron, L., Pagnoni, G., Drake, D. F., Woolwine, B. J., Spivey, J. R., Crowe, R. J., … Miller, A. H. (2012). Dopaminergic mechanisms of reduced basal ganglia responses to hedonic reward during interferon alfa administration. Archives of General Psychiatry, 69, 10441053. doi:10.1001/archgenpsychiatry.2011.2094CrossRefGoogle ScholarPubMed
Carpenter, L. L., Gawuga, C. E., Tyrka, A. R., Lee, J. K., Anderson, G. M., & Price, L. H. (2010). Association between plasma IL-6 response to acute stress and early-life adversity in healthy adults. Neuropsychopharmacology, 35, 26172623. doi:10.1038/npp.2010.159CrossRefGoogle ScholarPubMed
Carpenter, L. L., Gawuga, C. E., Tyrka, A. R., & Price, L. H. (2012). C-reactive protein, early life stress, and wellbeing in healthy adults. Acta Psychiatr Scand, 126, 402410. doi:10.1111/j.1600-0447.2012.01892.xCrossRefGoogle ScholarPubMed
Carroll, J. E., Gruenewald, T. L., Taylor, S. E., Janicki-Deverts, D., Matthews, K. A., & Seeman, T. E. (2013). Childhood abuse, parental warmth, and adult multisystem biological risk in the Coronary Artery Risk Development in Young Adults study. Proceedings of the National Academy of Sciences of the United States of America, 110, 1714917153. doi:10.1073/pnas.1315458110CrossRefGoogle ScholarPubMed
Chalasani, S. H., Sabelko, K. A., Sunshine, M. J., Littman, D. R., & Raper, J. A. (2003). A chemokine, SDF-1, reduces the effectiveness of multiple axonal repellents and is required for normal axon pathfinding. Journal of Neuroscience, 23, 13601371. doi:10.1523/jneurosci.23-04-01360.2003.CrossRefGoogle ScholarPubMed
Chen, E., Lee, W. K., Cavey, L., & Ho, A. (2013). Role models and the psychological characteristics that buffer low-socioeconomic-status youth from cardiovascular risk. Child Development, 84, 12411252. doi:10.1111/cdev.12037CrossRefGoogle ScholarPubMed
Chocyk, A., Dudys, D., Przyborowska, A., Majcher, I., Mackowiak, M., & Wedzony, K. (2011). Maternal separation affects the number, proliferation and apoptosis of glia cells in the substantia Nigra and ventral tegmental area of juvenile rats. Neuroscience, 173, 118. doi:10.1016/j.neuroscience.2010.11.037CrossRefGoogle ScholarPubMed
Chrysohoou, C., Panagiotakos, D. B., Pitsavos, C., Das, U. N., & Stefanadis, C. (2004). Adherence to the Mediterranean diet attenuates inflammation and coagulation process in healthy adults: The ATTICA Study. Journal of the American College of Cardiology, 44, 152158. doi:10.1016/j.jacc.2004.03.039CrossRefGoogle Scholar
Cicchetti, D., Handley, E. D., & Rogosch, F. A. (2015). Child maltreatment, inflammation, and internalizing symptoms: Investigating the roles of C-reactive protein, gene variation, and neuroendocrine regulation. Development and Psychopathology, 27, 553566. doi:10.1017/S0954579415000152CrossRefGoogle ScholarPubMed
Clarke, R., Shipley, M., Armitage, J., Collins, R., & Harris, W. (2009). Plasma phospholipid fatty acids and CHD in older men: Whitehall study of London civil servants. British Journal of Nutrition, 102, 279284. doi:10.1017/S0007114508143562CrossRefGoogle ScholarPubMed
Coe, C. L., Lubach, G. R., & Shirtcliff, E. A. (2007). Maternal stress during pregnancy predisposes for iron deficiency in infant monkeys impacting innate immunity. Pediatric Research, 61, 520524. doi:10.1203/pdr.0b013e318045be53CrossRefGoogle ScholarPubMed
Coe, C. L., Rosenberg, L. T., & Levine, S. (1988). Prolonged effect of psychological disturbance on macrophage chemiluminescence in the squirrel monkey. Brain Behavior & Immunity, 2, 151160. doi:10.1016/0889-1591(88)90015-3CrossRefGoogle ScholarPubMed
Cohen, S., Doyle, W. J., Alper, C. M., Janicki-Deverts, D., & Turner, R. B. (2009). Sleep habits and susceptibility to the common cold. Archives of Internal Medicine, 169, 6267. doi:10.1001/archinternmed.2008.505CrossRefGoogle ScholarPubMed
Cohen, S., Janicki-Deverts, D., Doyle, W. J., Miller, G. E., Frank, E., Rabin, B. S., & Turner, R. B. (2012). Chronic stress, glucocorticoid receptor resistance, inflammation, and disease risk. Proceedings of the National Acadamy of Sciences of the United States of America, 109, 59955999. doi:10.1073/pnas.1118355109CrossRefGoogle ScholarPubMed
Cole, S. W., Conti, G., Arevalo, J. M., Ruggiero, A. M., Heckman, J. J., & Suomi, S. J. (2012). Transcriptional modulation of the developing immune system by early life social adversity. Proceedings of the National Acadamy of Sciences of the United States of America, 109, 2057820583. doi:10.1073/pnas.1218253109CrossRefGoogle ScholarPubMed
Conn, V. S. (2010). Depressive symptom outcomes of physical activity interventions: Meta-analysis findings. Annals of Behavioral Medicine, 39, 128138. doi:10.1007/s12160-010-9172-xCrossRefGoogle ScholarPubMed
Copeland, W. E., Wolke, D., Lereya, S. T., Shanahan, L., Worthman, C., & Costello, E. J. (2014). Childhood bullying involvement predicts low-grade systemic inflammation into adulthood. Proceedings of the National Acadamy of Sciences of the United States of America, 111, 75707575. doi:10.1073/pnas.1323641111CrossRefGoogle ScholarPubMed
Creswell, J. D., Irwin, M. R., Burklund, L. J., Lieberman, M. D., Arevalo, J. M., Ma, J., … Cole, S. W. (2012). Mindfulness-based stress reduction training reduces loneliness and pro-inflammatory gene expression in older adults: A small randomized controlled trial. Brain Behavior & Immunity, 26, 10951101. doi:10.1016/j.bbi.2012.07.006CrossRefGoogle ScholarPubMed
Cryan, J. F., & Dinan, T. G. (2012). Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nature Reviews Neuroscience, 13, 701712. doi:10.1038/nrn3346CrossRefGoogle ScholarPubMed
Cryan, J. F., & Holmes, A. (2005). The ascent of mouse: Advances in modelling human depression and anxiety. Nature Rev Drug Discov, 4, 775790. doi:10.1038/nrd1825CrossRefGoogle ScholarPubMed
Dallman, M. F. (2010). Stress-induced obesity and the emotional nervous system. Trends in Endocrinology & Metabolism, 21, 159165. doi:10.1016/j.tem.2009.10.004CrossRefGoogle ScholarPubMed
Dallman, M. F., Pecoraro, N. C., & la Fleur, S. E. (2005). Chronic stress and comfort foods: Self-medication and abdominal obesity. Brain Behavior & Immunity, 19, 275280. doi:10.1016/j.bbi.2004.11.004CrossRefGoogle ScholarPubMed
Dallman, M. F., Strack, A. M., Akana, S. F., Bradbury, M. J., Hanson, E. S., Scribner, K. A., & Smith, M. (1993). Feast and famine: Critical role of glucocorticoids with insulin in daily energy flow. Frontiers in Neuroendocrinology, 14, 303347. doi:10.1006/frne.1993.1010CrossRefGoogle ScholarPubMed
Danese, A. (2020). Annual Research Review: Rethinking childhood trauma-new research directions for measurement, study design and analytical strategies. Journal of Child Psychology & Psychiatry, 61, 236250. doi:10.1111/jcpp.13160CrossRefGoogle ScholarPubMed
Danese, A., & Baldwin, J. R. (2017). Hidden wounds? Inflammatory links between childhood trauma and psychopathology. Annual Review of Psychology, 68, 517544. doi:10.1146/annurev-psych-010416-044208CrossRefGoogle ScholarPubMed
Danese, A., Caspi, A., Williams, B., Ambler, A., Sugden, K., Mika, J., … Arseneault, L. (2011). Biological embedding of stress through inflammation processes in childhood. Molecular Psychiatry, 16, 244246. doi:10.1038/mp.2010.5CrossRefGoogle ScholarPubMed
Danese, A., Dove, R., Belsky, D. W., Henchy, J., Williams, B., Ambler, A., & Arseneault, L. (2014). Leptin deficiency in maltreated children. Translational Psychiatry, 4, e446. doi:10.1038/tp.2014.79CrossRefGoogle ScholarPubMed
Danese, A., & Lewis, J. S. (2017). Psychoneuroimmunology of early-life stress: The hidden wounds of childhood trauma? Neuropsychopharmacology, 42, 99114. doi:10.1038/npp.2016.198CrossRefGoogle ScholarPubMed
Danese, A., & McEwen, B. S. (2012). Adverse childhood experiences, allostasis, allostatic load, and age-related disease. Physiology & Behavior, 106, 2939. doi:10.1016/j.physbeh.2011.08.019CrossRefGoogle ScholarPubMed
Danese, A., Moffitt, T. E., Pariante, C. M., Ambler, A., Poulton, R., & Caspi, A. (2008). Elevated inflammation levels in depressed adults with a history of childhood maltreatment. Archives of General Psychiatry, 65, 409415. doi:10.1001/archpsyc.65.4.409CrossRefGoogle ScholarPubMed
Danese, A., Pariante, C. M., Caspi, A., Taylor, A., & Poulton, R. (2007). Childhood maltreatment predicts adult inflammation in a life-course study. Proceedings of the National Academy of Sciences of the United States of America, 104, 13191324. doi:10.1073/pnas.0610362104CrossRefGoogle Scholar
Danese, A., & Tan, M. (2014). Childhood maltreatment and obesity: Systematic review and meta-analysis. Molecular Psychiatry, 19, 544554. doi:10.1038/mp.2013.54CrossRefGoogle ScholarPubMed
Dantzer, R., O'Connor, J. C., Freund, G. G., Johnson, R. W., & Kelley, K. W. (2008). From inflammation to sickness and depression: When the immune system subjugates the brain. Nature Reviews Neuroscience, 9, nrn2297. doi:10.1038/nrn2297CrossRefGoogle ScholarPubMed
Dargel, A. A., Godin, O., Kapczinski, F., Kupfer, D. J., & Leboyer, M. (2015). C-reactive protein alterations in bipolar disorder: A meta-analysis. Journal of Clinical Psychiatry, 76, 142150. doi:10.4088/JCP.14r09007CrossRefGoogle ScholarPubMed
David, J., Measelle, J., Ostlund, B., & Ablow, J. (2017). Association between early life adversity and inflammation during infancy. Developmental Psychobiology, 59, 696702. doi:10.1002/dev.21538CrossRefGoogle ScholarPubMed
Del Giudice, M., & Gangestad, S. W. (2018). Rethinking IL-6 and CRP: Why they are more than inflammatory biomarkers, and why it matters. Brain Behavior & Immunity, 70, 6175. doi:10.1016/j.bbi.2018.02.013CrossRefGoogle ScholarPubMed
Dennison, U., McKernan, D., Cryan, J., & Dinan, T. (2012). Schizophrenia patients with a history of childhood trauma have a pro-inflammatory phenotype. Psychological Medicine, 42, 18651871. doi:10.1017/S0033291712000074CrossRefGoogle ScholarPubMed
Deverman, B. E., & Patterson, P. H. (2009). Cytokines and CNS development. Neuron, 64, 6178. doi:10.1016/j.neuron.2009.09.002CrossRefGoogle ScholarPubMed
Deykin, E. Y., & MacMahon, B. (1979). Viral exposure and autism. American Journal of Epidemiology, 109, 628638. doi:10.1093/oxfordjournals.aje.a112726CrossRefGoogle ScholarPubMed
Dillon, D. G., Holmes, A. J., Birk, J. L., Brooks, N., Lyons-Ruth, K., & Pizzagalli, D. A. (2009). Childhood adversity is associated with left basal ganglia dysfunction during reward anticipation in adulthood. Biological Psychiatry, 66, 206213. doi:10.1016/j.biopsych.2009.02.019CrossRefGoogle ScholarPubMed
Di Nicola, M., Cattaneo, A., Hepgul, N., Di Forti, M., Aitchison, K. J., Janiri, L., … Mondelli, V. (2013). Serum and gene expression profile of cytokines in first-episode psychosis. Brain Behavior & Immunity, 31, 9095. doi:10.1016/j.bbi.2012.06.010CrossRefGoogle ScholarPubMed
Dube, S. R., Felitti, V. J., Dong, M., Chapman, D. P., Giles, W. H., & Anda, R. F. (2003). Childhood abuse, neglect, and household dysfunction and the risk of illicit drug use: The adverse childhood experiences study. Pediatrics, 111, 564572. doi:10.1542/peds.111.3.564CrossRefGoogle ScholarPubMed
Dunn, A. J. (2000). Cytokine activation of the HPA axis. Annals of the New York Academy of Sciences, 917, 608617. doi:10.1111/j.1749-6632.2000.tb05426.xCrossRefGoogle ScholarPubMed
Eisenberger, N. I., Berkman, E. T., Inagaki, T. K., Rameson, L. T., Mashal, N. M., & Irwin, M. R. (2010). Inflammation-induced anhedonia: Endotoxin reduces ventral striatum responses to reward. Biological Psychiatry, 68, 748754. doi:10.1016/j.biopsych.2010.06.010CrossRefGoogle Scholar
Ekdahl, C. T., Claasen, J.-H., Bonde, S., Kokaia, Z., & Lindvall, O. (2003). Inflammation is detrimental for neurogenesis in adult brain. Proceedings of the National Academy of Sciences, 100, 1363213637. doi:10.1073/pnas.2234031100CrossRefGoogle ScholarPubMed
Elwenspoek, M. M. C., Hengesch, X., Leenen, F. A. D., Schritz, A., Sias, K., Schaan, V. K., … Muller, C. P. (2017a). Proinflammatory T cell status associated with early life adversity. Journal of Immunology, 199, 40464055. doi:10.4049/jimmunol.1701082CrossRefGoogle Scholar
Elwenspoek, M. M. C., Sias, K., Hengesch, X., Schaan, V. K., Leenen, F. A. D., Adams, P., … Turner, J. D. (2017b). T cell immunosenescence after early life adversity: Association with cytomegalovirus infection. Frontiers in Immunology, 8, 1263. doi:10.3389/fimmu.2017.01263CrossRefGoogle Scholar
Enache, D., Pariante, C. M., & Mondelli, V. (2019). Markers of central inflammation in major depressive disorder: A systematic review and meta-analysis of studies examining cerebrospinal fluid, positron emission tomography and post-mortem brain tissue. Brain Behavior & Immunity, 81, 2440. doi:10.1016/j.bbi.2019.06.015CrossRefGoogle ScholarPubMed
Eraly, S. A., Nievergelt, C. M., Maihofer, A. X., Barkauskas, D. A., Biswas, N., Agorastos, A., … Marine Resiliency Study, T. (2014). Assessment of plasma C-reactive protein as a biomarker of posttraumatic stress disorder risk. JAMA Psychiatry, 71, 423431. doi:10.1001/jamapsychiatry.2013.4374CrossRefGoogle ScholarPubMed
Esposito, K., Marfella, R., Ciotola, M., Di Palo, C., Giugliano, F., Giugliano, G., … Giugliano, D. (2004). Effect of a Mediterranean-style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: A randomized trial. JAMA, 292, 14401446. doi:10.1001/jama.292.12.1440CrossRefGoogle ScholarPubMed
Evans, G. W., Fuller-Rowell, T. E., & Doan, S. N. (2012). Childhood cumulative risk and obesity: The mediating role of self-regulatory ability. Pediatrics, 129, e6873. doi:10.1542/peds.2010-3647CrossRefGoogle ScholarPubMed
Fedewa, M. V., Hathaway, E. D., & Ward-Ritacco, C. L. (2017). Effect of exercise training on C reactive protein: A systematic review and meta-analysis of randomised and non-randomised controlled trials. British Journal of Sports Medicine, 51, 670676. doi:10.1136/bjsports-2016-095999CrossRefGoogle Scholar
Flouri, E., Francesconi, M., Midouhas, E., & Lewis, G. (2020). Prenatal and childhood adverse life events, inflammation and depressive symptoms across adolescence. Journal of Affective Disorders, 260, 577582. doi:10.1016/j.jad.2019.09.024CrossRefGoogle ScholarPubMed
Flouri, E., Francesconi, M., Papachristou, E., Midouhas, E., & Lewis, G. (2019). Stressful life events, inflammation and emotional and behavioural problems in children: A population-based study. Brain Behavior & Immunity, 80, 6672. doi:10.1016/j.bbi.2019.02.023CrossRefGoogle ScholarPubMed
Ford, D. E., & Erlinger, T. P. (2004). Depression and C-reactive protein in US adults: Data from the Third National Health and Nutrition Examination Survey. Archives of Internal Medicine, 164, 10101014. doi:10.1001/archinte.164.9.1010CrossRefGoogle ScholarPubMed
Francis, D., Diorio, J., Liu, D., & Meaney, M. J. (1999). Nongenomic transmission across generations of maternal behavior and stress responses in the rat. Science, 286, 11551158. doi:10.1126/science.286.5442.1155CrossRefGoogle ScholarPubMed
Frank, P., Kaushal, A., Poole, L., Lawes, S., Chalder, T., & Cadar, D. (2019). Systemic low-grade inflammation and subsequent depressive symptoms: Is there a mediating role of physical activity? Brain Behavior & Immunity, 80, 688696. doi:10.1016/j.bbi.2019.05.017CrossRefGoogle Scholar
Frank, M. G., Watkins, L. R., & Maier, S. F. (2013). Stress-induced glucocorticoids as a neuroendocrine alarm signal of danger. Brain Behavior & Immunity, 33, 16. doi:10.1016/j.bbi.2013.02.004CrossRefGoogle ScholarPubMed
Frodl, T., Carballedo, A., Hughes, M. M., Saleh, K., Fagan, A., Skokauskas, N., … Connor, T. J. (2012). Reduced expression of glucocorticoid-inducible genes GILZ and SGK-1: High IL-6 levels are associated with reduced hippocampal volumes in major depressive disorder. Translational Psychiatry, 2, e88. doi:10.1038/tp.2012.14.CrossRefGoogle ScholarPubMed
Fung, T. T., Rimm, E. B., Spiegelman, D., Rifai, N., Tofler, G. H., Willett, W. C., & Hu, F. B. (2001). Association between dietary patterns and plasma biomarkers of obesity and cardiovascular disease risk. The American Journal of Clinical Nutrition, 73, 6167. doi:10.1093/ajcn/73.1.61CrossRefGoogle ScholarPubMed
Ganguly, P., & Brenhouse, H. C. (2015). Broken or maladaptive? Altered trajectories in neuroinflammation and behavior after early life adversity. Developmental Cognitive Neuroscience, 11, 1830. doi:10.1016/j.dcn.2014.07.001CrossRefGoogle ScholarPubMed
Garcia-Rodenas, C. L., Bergonzelli, G. E., Nutten, S., Schumann, A., Cherbut, C., Turini, M., … Corthesy-Theulaz, I. (2006). Nutritional approach to restore impaired intestinal barrier function and growth after neonatal stress in rats. Journal of Pediatric Gastroenterology Nutrition, 43, 1624. doi:10.1097/01.mpg.0000226376.95623.9fCrossRefGoogle ScholarPubMed
Gareau, M. G., Silva, M. A., & Perdue, M. H. (2008). Pathophysiological mechanisms of stress-induced intestinal damage. Currrent Molecular Medicine, 8, 274281. doi:10.2174/156652408784533760CrossRefGoogle ScholarPubMed
Gilbert, R., Widom, C. S., Browne, K., Fergusson, D., Webb, E., & Janson, S. (2009). Burden and consequences of child maltreatment in high-income countries. Lancet, 373, 6881. doi:10.1016/S0140-6736(08)61706-7CrossRefGoogle ScholarPubMed
Goosby, B. J., Malone, S., Richardson, E. A., Cheadle, J. E., & Williams, D. T. (2015). Perceived discrimination and markers of cardiovascular risk among low-income African American youth. American Journal of Human Biology, 27, 546552. doi:10.1002/ajhb.22683CrossRefGoogle ScholarPubMed
Gouin, J. P., Glaser, R., Malarkey, W. B., Beversdorf, D., & Kiecolt-Glaser, J. K. (2012). Childhood abuse and inflammatory responses to daily stressors. Annals of Behavioral Medicine, 44, 287292. doi:10.1007/s12160-012-9386-1CrossRefGoogle ScholarPubMed
Green, J. G., McLaughlin, K. A., Berglund, P. A., Gruber, M. J., Sampson, N. A., Zaslavsky, A. M., & Kessler, R. C. (2010). Childhood adversities and adult psychiatric disorders in the national comorbidity survey replication I: Associations with first onset of DSM-IV disorders. Archives of General Psychiatry, 67, 113123. doi:10.1001/archgenpsychiatry.2009.186CrossRefGoogle ScholarPubMed
Gregory, A. M., & Sadeh, A. (2016). Annual Research Review: Sleep problems in childhood psychiatric disorders--a review of the latest science. Journal of Child Psychology and Psychiatry, 57, 296317. doi:10.1111/jcpp.12469CrossRefGoogle ScholarPubMed
Gundersen, C., Mahatmya, D., Garasky, S., & Lohman, B. (2011). Linking psychosocial stressors and childhood obesity. Obesity Reviews, 12, e5463. doi:10.1111/j.1467-789X.2010.00813.xCrossRefGoogle ScholarPubMed
Gunnar, M. R., & Reid, B. M. (2019). Early deprivation revisited: Contemporary studies of the impact on young children of institutional care. Annual Review of Developmental Psychology, 1, 93118. doi:10.1146/annurev-devpsych-121318-085013CrossRefGoogle Scholar
Hadley, C., & Decaro, J. A. (2014). Testing hypothesized predictors of immune activation in Tanzanian infants and children: Community, household, caretaker, and child effects. American Journal of Human Biology, 26, 523529. doi:10.1002/ajhb.22558CrossRefGoogle ScholarPubMed
Hanisch, U. K., & Kettenmann, H. (2007). Microglia: Active sensor and versatile effector cells in the normal and pathologic brain. Nature Neuroscience, 10, 13871394. doi:10.1038/nn1997CrossRefGoogle ScholarPubMed
Harlow, H. F., Dodsworth, R. O., & Harlow, M. K. (1965). Total social isolation in monkeys. Proceedings of the National Academy of Sciences of the United States of America, 54, 9097. doi:10.1073/pnas.54.1.90CrossRefGoogle ScholarPubMed
Harrison, N. A., Brydon, L., Walker, C., Gray, M. A., Steptoe, A., & Critchley, H. D. (2009). Inflammation causes mood changes through alterations in subgenual cingulate activity and mesolimbic connectivity. Biological Psychiatry, 66, 407414. doi:10.1016/j.biopsych.2009.03.015CrossRefGoogle ScholarPubMed
Hartwell, K. J., Moran-Santa Maria, M. M., Twal, W. O., Shaftman, S., DeSantis, S. M., McRae-Clark, A. L., & Brady, K. T. (2013). Association of elevated cytokines with childhood adversity in a sample of healthy adults. Journal of Psychiatric Research, 47, 604610. doi:10.1016/j.jpsychires.2013.01.008CrossRefGoogle Scholar
Heim, C., Ehlert, U., & Hellhammer, D. H. (2000). The potential role of hypocortisolism in the pathophysiology of stress-related bodily disorders. Psychoneuroendocrinology, 25, 135. doi:10.1016/s0306-4530(99)00035-9CrossRefGoogle ScholarPubMed
Hemmings, S. M. J., Malan-Muller, S., van den Heuvel, L. L., Demmitt, B. A., Stanislawski, M. A., Smith, D. G., … Lowry, C. A. (2017). The microbiome in posttraumatic stress disorder and trauma-exposed controls: An exploratory study. Psychosomatic Medicine, 79, 936946. doi:10.1097/PSY.0000000000000512CrossRefGoogle ScholarPubMed
Hennessy, M. B., Deak, T., & Schiml-Webb, P. A. (2010a). Early attachment-figure separation and increased risk for later depression: Potential mediation by proinflammatory processes. Neuroscience & Biobehavioral Reviews, 34, 782790. doi:10.1016/j.neubiorev.2009.03.012CrossRefGoogle Scholar
Hennessy, M. B., Deak, T., Schiml-Webb, P. A., Carlisle, C. W., & O'Brien, E. (2010b). Maternal separation produces, and a second separation enhances, core temperature and passive behavioral responses in guinea pig pups. Physiology & Behavior, 100, 305310. doi:10.1016/j.physbeh.2010.02.024CrossRefGoogle Scholar
Hepgul, N., Pariante, C. M., Dipasquale, S., DiForti, M., Taylor, H., Marques, T. R., … Mondelli, V. (2012). Childhood maltreatment is associated with increased body mass index and increased C-reactive protein levels in first-episode psychosis patients. Psychological Medicine, 42, 18931901. doi:10.1017/S0033291711002947CrossRefGoogle ScholarPubMed
Hinde, R. A., & Spencer-Booth, Y. (1971). Effects of brief separation from mother on rhesus monkeys. Science, 173, 111118. doi:10.1126/science.173.3992.111CrossRefGoogle ScholarPubMed
Hooper, L. V., Littman, D. R., & Macpherson, A. J. (2012). Interactions between the microbiota and the immune system. Science, 336, 12681273. doi:10.1126/science.1223490CrossRefGoogle ScholarPubMed
Hostinar, C. E., Lachman, M. E., Mroczek, D. K., Seeman, T. E., & Miller, G. E. (2015a). Additive contributions of childhood adversity and recent stressors to inflammation at midlife: Findings from the MIDUS study. Developmental Psychology, 51, 1630. doi:10.1037/dev0000049CrossRefGoogle Scholar
Hostinar, C. E., Ross, K. M., Chen, E., & Miller, G. E. (2015b). Modeling the association between lifecourse socioeconomic disadvantage and systemic inflammation in healthy adults: The role of self-control. Health Psychology, 34, 580590. doi:10.1037/hea0000130CrossRefGoogle Scholar
Howren, M. B., Lamkin, D. M., & Suls, J. (2009). Associations of depression with C-reactive protein, IL-1, and IL-6: A meta-analysis. Psychosomatic Medicine, 71, 171186. doi:10.1097/PSY.0b013e3181907c1bCrossRefGoogle ScholarPubMed
Huh, G. S., Boulanger, L. M., Du, H., Riquelme, P. A., Brotz, T. M., & Shatz, C. J. (2000). Functional requirement for class I MHC in CNS development and plasticity. Science, 290, 21552159. doi:10.1126/science.290.5499.2155CrossRefGoogle ScholarPubMed
Humphreys, K. L., & Zeanah, C. H. (2015). Deviations from the expectable environment in early childhood and emerging psychopathology. Neuropsychopharmacology, 40, 154170. doi:10.1038/npp.2014.165CrossRefGoogle ScholarPubMed
Inagaki, T. K., Muscatell, K. A., Irwin, M. R., Cole, S. W., & Eisenberger, N. I. (2012). Inflammation selectively enhances amygdala activity to socially threatening images. NeuroImage, 59, 32223226. doi:10.1016/j.neuroimage.2011.10.090CrossRefGoogle ScholarPubMed
Irwin, M. R. (2015). Why sleep is important for health: A psychoneuroimmunology perspective. Annual Review of Psychology, 66, 143172. doi:10.1146/annurev-psych-010213-115205CrossRefGoogle ScholarPubMed
Irwin, M. R., & Cole, S. W. (2011). Reciprocal regulation of the neural and innate immune systems. Nature Reviews Immunology, 11, 625632. doi:10.1038/nri3042CrossRefGoogle ScholarPubMed
Irwin, M. R., & Olmstead, R. (2012). Mitigating cellular inflammation in older adults: A randomized controlled trial of Tai Chi Chih. American Journal of Geriatric Psychiatry, 20, 764772. doi:10.1097/JGP.0b013e3182330fd3CrossRefGoogle ScholarPubMed
Irwin, M. R., Olmstead, R., Carrillo, C., Sadeghi, N., Breen, E. C., Witarama, T., … Nicassio, P. (2014). Cognitive behavioral therapy vs. Tai Chi for late life insomnia and inflammatory risk: A randomized controlled comparative efficacy trial. Sleep, 37, 15431552. doi:10.5665/sleep.4008CrossRefGoogle ScholarPubMed
Irwin, M. R., Olmstead, R., & Motivala, S. J. (2008). Improving sleep quality in older adults with moderate sleep complaints: A randomized controlled trial of Tai Chi Chih. Sleep, 31, 10011008. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/18652095Google ScholarPubMed
Irwin, M. R., Olmstead, R., & Oxman, M. N. (2007). Augmenting immune responses to varicella zoster virus in older adults: A randomized, controlled trial of Tai Chi. Journal of the American Geriatrics Society, 55, 511517. doi:10.1111/j.1532-5415.2007.01109.xCrossRefGoogle ScholarPubMed
Irwin, M. R., Wang, M., Campomayor, C. O., Collado-Hidalgo, A., & Cole, S. (2006). Sleep deprivation and activation of morning levels of cellular and genomic markers of inflammation. Archives of Internal Medicine, 166, 17561762. doi:10.1001/archinte.166.16.1756CrossRefGoogle ScholarPubMed
Iwata, N. G., Pham, M., Rizzo, N. O., Cheng, A. M., Maloney, E., & Kim, F. (2011). Trans fatty acids induce vascular inflammation and reduce vascular nitric oxide production in endothelial cells. PLoS One, 6, e29600. doi:10.1371/journal.pone.0029600CrossRefGoogle ScholarPubMed
Jacka, F. N., Kremer, P. J., Berk, M., de Silva-Sanigorski, A. M., Moodie, M., Leslie, E. R., … Swinburn, B. A. (2011a). A prospective study of diet quality and mental health in adolescents. PLoS One, 6, e24805. doi:10.1371/journal.pone.0024805CrossRefGoogle Scholar
Jacka, F. N., Kremer, P. J., Leslie, E. R., Berk, M., Patton, G. C., Toumbourou, J. W., & Williams, J. W. (2009). Associations between diet quality and depressed mood in adolescents: Results from the Australian healthy neighbourhoods study. Australian and New Zealand Journal of Psychiatry, 44, 435442. doi:10.3109/00048670903571598CrossRefGoogle Scholar
Jacka, F. N., Mykletun, A., Berk, M., Bjelland, I., & Tell, G. S. (2011b). The association between habitual diet quality and the common mental disorders in community-dwelling adults. Psychosomatic Medicine, 73, 483490. doi:10.1097/psy.0b013e318222831aCrossRefGoogle Scholar
Jacka, F. N., Pasco, J. A., Williams, L. J., Leslie, E. R., Dodd, S., Nicholson, G. C., … Berk, M. (2011c). Lower levels of physical activity in childhood associated with adult depression. Journal of Science and Medicine in Sport, 14, 222226. doi:10.1016/j.jsams.2010.10.458CrossRefGoogle Scholar
Jacka, F. N., Pasco, J. A., Williams, L. J., Meyer, B. J., Digger, R., & Berk, M. (2013). Dietary intake of fish and PUFA, and clinical depressive and anxiety disorders in women. British Journal of Nutrition, 109, 20592066. doi:10.1017/S0007114512004102CrossRefGoogle ScholarPubMed
Jasarevic, E., Howard, C. D., Misic, A. M., Beiting, D. P., & Bale, T. L. (2017). Stress during pregnancy alters temporal and spatial dynamics of the maternal and offspring microbiome in a sex-specific manner. Scientific Reports, 7, 44182. doi:10.1038/srep44182CrossRefGoogle Scholar
Jasarevic, E., Rodgers, A. B., & Bale, T. L. (2015). A novel role for maternal stress and microbial transmission in early life programming and neurodevelopment. Neurobiology of Stress, 1, 8188. doi:10.1016/j.ynstr.2014.10.005CrossRefGoogle ScholarPubMed
Jelleyman, T., & Spencer, N. (2008). Residential mobility in childhood and health outcomes: A systematic review. Journal of Epidemiology and Community Health, 62, 584592. doi:10.1136/jech.2007.060103CrossRefGoogle ScholarPubMed
Johansen, J. P., Cain, C. K., Ostroff, L. E., & LeDoux, J. E. (2011). Molecular mechanisms of fear learning and memory. Cell, 147, 509524. doi:10.1016/j.cell.2011.10.009CrossRefGoogle ScholarPubMed
John-Henderson, N. A., Henderson-Matthews, B., Ollinger, S. R., Racine, J., Gordon, M. R., Higgins, A. A., … Rynda-Apple, A. (2020). Adverse childhood experiences and immune system inflammation in adults residing on the blackfeet reservation: The moderating role of sense of belonging to the community. Annals of Behavioral Medicine, 54, 8793. doi:10.1093/abm/kaz029CrossRefGoogle ScholarPubMed
Kessler, R. C., Davis, C. G., & Kendler, K. S. (1997). Childhood adversity and adult psychiatric disorder in the US National Comorbidity Survey. Psychological Medicine, 27, 11011119. doi:10.1017/s0033291797005588CrossRefGoogle ScholarPubMed
Kessler, R. C., McLaughlin, K. A., Green, J. G., Gruber, M. J., Sampson, N. A., Zaslavsky, A. M., … Williams, D. R. (2010). Childhood adversities and adult psychopathology in the WHO World Mental Health Surveys. British Journal of Psychiatry, 197, 378385. doi:10.1192/bjp.bp.110.080499CrossRefGoogle ScholarPubMed
Khandaker, G. M., Pearson, R. M., Zammit, S., Lewis, G., & Jones, P. B. (2014). Association of serum interleukin 6 and C-reactive protein in childhood with depression and psychosis in young adult life: A population-based longitudinal study. JAMA Psychiatry, 71, 11211128. doi:10.1001/jamapsychiatry.2014.1332CrossRefGoogle ScholarPubMed
Khandaker, G. M., Zimbron, J., Lewis, G., & Jones, P. B. (2013). Prenatal maternal infection, neurodevelopment and adult schizophrenia: A systematic review of population-based studies. Psychological Medicine, 43, 239257. doi:10.1017/S0033291712000736CrossRefGoogle ScholarPubMed
Kiecolt-Glaser, J. K., Gouin, J. P., Weng, N. P., Malarkey, W. B., Beversdorf, D. Q., & Glaser, R. (2011). Childhood adversity heightens the impact of later-life caregiving stress on telomere length and inflammation. Psychosomatic Medicine, 73, 1622. doi:10.1097/PSY.0b013e31820573b6CrossRefGoogle ScholarPubMed
Kilic, S., Ozlem, K., & Murat, T. (2017). Inflammatory parameters in sexually abused children. Saudi Medical Journal, 38, 12131218. doi:10.15537/smj.2017.12.21463Google ScholarPubMed
King, D. E., Mainous, A. G., 3rd, Geesey, M. E., & Woolson, R. F. (2005). Dietary magnesium and C-reactive protein levels. Journal of the American College of Nutrition, 24, 166171. doi:10.1080/07315724.2005.10719461CrossRefGoogle ScholarPubMed
Kipnis, J., Gadani, S., & Derecki, N. C. (2012). Pro-cognitive properties of T cells. Nature Reviews Immunology, 12, 663669. doi:10.1038/nri3280CrossRefGoogle ScholarPubMed
Klengel, T., Mehta, D., Anacker, C., Rex-Haffner, M., Pruessner, J. C., Pariante, C. M., … Binder, E. B. (2013). Allele-specific FKBP5 DNA demethylation mediates gene-childhood trauma interactions. Nature Neuroscience, 16, 3341. doi:10.1038/nn.3275CrossRefGoogle ScholarPubMed
Knuesel, I., Chicha, L., Britschgi, M., Schobel, S. A., Bodmer, M., Hellings, J. A., … Prinssen, E. P. (2014). Maternal immune activation and abnormal brain development across CNS disorders. Nature Reviews Neurology, 10, 643660. doi:10.1038/nrneurol.2014.187CrossRefGoogle ScholarPubMed
Kohlboeck, G., Sausenthaler, S., Standl, M., Koletzko, S., Bauer, C. P., von Berg, A., … Groups, L. I.-p. S. (2012). Food intake, diet quality and behavioral problems in children: Results from the GINI-plus/LISA-plus studies. Annals of Nutrition and Metabolism, 60, 247256. doi:10.1159/000337552CrossRefGoogle ScholarPubMed
Kohler, O., Benros, M. E., Nordentoft, M., Farkouh, M. E., Iyengar, R. L., Mors, O., & Krogh, J. (2014). Effect of anti-inflammatory treatment on depression, depressive symptoms, and adverse effects: A systematic review and meta-analysis of randomized clinical trials. JAMA Psychiatry, 71, 13811391. doi:10.1001/jamapsychiatry.2014.1611CrossRefGoogle ScholarPubMed
Konturek, P. C., Brzozowski, T., & Konturek, S. J. (2011). Stress and the gut: Pathophysiology, clinical consequences, diagnostic approach and treatment options. Journal of Physiology and Pharmacology, 62, 591599. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/22314561Google ScholarPubMed
Koss, K. J., & Gunnar, M. R. (2018). Annual Research Review: Early adversity, the hypothalamic-pituitary-adrenocortical axis, and child psychopathology. Journal of Child Psychology and Psychiatry, 59, 327346. doi:10.1111/jcpp.12784CrossRefGoogle ScholarPubMed
Koss, K. J., Hostinar, C. E., Donzella, B., & Gunnar, M. R. (2014). Social deprivation and the HPA axis in early development. Psychoneuroendocrinology, 50, 113. doi:10.1016/j.psyneuen.2014.07.028CrossRefGoogle ScholarPubMed
Koss, K. J., Mliner, S. B., Donzella, B., & Gunnar, M. R. (2016). Early adversity, hypocortisolism, and behavior problems at school entry: A study of internationally adopted children. Psychoneuroendocrinology, 66, 3138.CrossRefGoogle ScholarPubMed
Kraynak, T. E., Marsland, A. L., Hanson, J. L., & Gianaros, P. J. (2019). Retrospectively reported childhood physical abuse, systemic inflammation, and resting corticolimbic connectivity in midlife adults. Brain Behavior & Immunity, 82, 203213. doi:10.1016/j.bbi.2019.08.186CrossRefGoogle ScholarPubMed
Kuczmarski, M. F., Cremer Sees, A., Hotchkiss, L., Cotugna, N., Evans, M. K., & Zonderman, A. B. (2010). Higher Healthy Eating Index-2005 scores associated with reduced symptoms of depression in an urban population: Findings from the Healthy Aging in Neighborhoods of Diversity Across the Life Span (HANDLS) study. Journal of the American Dietetic Association, 110, 383389. doi:10.1016/j.jada.2009.11.025CrossRefGoogle Scholar
Kuhlman, K. R., Horn, S. R., Chiang, J. J., & Bower, J. E. (2020). Early life adversity exposure and circulating markers of inflammation in children and adolescents: A systematic review and meta-analysis. Brain Behavior & Immunity, 86, 3042. doi:10.1016/j.bbi.2019.04.028CrossRefGoogle Scholar
Leboyer, M., Soreca, I., Scott, J., Frye, M., Henry, C., Tamouza, R., & Kupfer, D. J. (2012). Can bipolar disorder be viewed as a multi-system inflammatory disease? Journal of Affective Disorders, 141, 110. doi:10.1016/j.jad.2011.12.049CrossRefGoogle ScholarPubMed
LeDoux, J. (2003). The emotional brain, fear, and the amygdala. Cellular and Molecular Neurobiology, 23, 727738. doi:10.1023/a:1025048802629CrossRefGoogle ScholarPubMed
Leppanen, J. M., & Nelson, C. A. (2009). Tuning the developing brain to social signals of emotions. Nature Reviews Neuroscience, 10, 3747. doi:10.1038/nrn2554CrossRefGoogle ScholarPubMed
Levine, S., Alpert, M., & Lewis, G. W. (1957). Infantile experience and the maturation of the pituitary adrenal axis. Science, 126, 1347. doi:10.1126/science.126.3287.1347CrossRefGoogle ScholarPubMed
Levine, S., Chevalier, J. A., & Korchin, S. J. (1956). The effects of early shock and handling on later avoidance learning. Journal of Personality, 24, 475493. doi:10.1111/j.1467-6494.1956.tb01283.xCrossRefGoogle ScholarPubMed
Levitan, E. B., Cook, N. R., Stampfer, M. J., Ridker, P. M., Rexrode, K. M., Buring, J. E., … Liu, S. (2008). Dietary glycemic index, dietary glycemic load, blood lipids, and C-reactive protein. Metabolism, 57, 437443. doi:10.1016/j.metabol.2007.11.002CrossRefGoogle ScholarPubMed
Lieberam, I., Agalliu, D., Nagasawa, T., Ericson, J., & Jessell, T. M. (2005). A Cxcl12-CXCR4 chemokine signaling pathway defines the initial trajectory of mammalian motor axons. Neuron, 47, 667679. doi:10.1016/j.neuron.2005.08.011CrossRefGoogle ScholarPubMed
Liu, S., Manson, J. E., Buring, J. E., Stampfer, M. J., Willett, W. C., & Ridker, P. M. (2002). Relation between a diet with a high glycemic load and plasma concentrations of high-sensitivity C-reactive protein in middle-aged women. The American Journal of Clinical Nutrition, 75, 492498. doi:10.1093/ajcn/75.3.492CrossRefGoogle ScholarPubMed
Lopez-Garcia, E., Schulze, M. B., Fung, T. T., Meigs, J. B., Rifai, N., Manson, J. E., & Hu, F. B. (2004). Major dietary patterns are related to plasma concentrations of markers of inflammation and endothelial dysfunction. American Journal of Clinical Nutrition, 80, 10291035. doi:10.1093/ajcn/80.4.1029CrossRefGoogle Scholar
Lovallo, W. R. (2013). Early life adversity reduces stress reactivity and enhances impulsive behavior: Implications for health behaviors. International Journal of Psychophysiology, 90, 816. doi:10.1016/j.ijpsycho.2012.10.006CrossRefGoogle ScholarPubMed
Lu, S., Peng, H., Wang, L., Vasisha, S., Zhang, Y., Gao, W., … Li, L. (2013). Elevated specific peripheral cytokines found in major depressive disorder patients with childhood trauma exposure: A cytokine antibody array analysis. Comprehensive Psychiatry, 54, 953961. doi:10.1016/j.comppsych.2013.03.026CrossRefGoogle ScholarPubMed
Mackos, A. R., Maltz, R., & Bailey, M. T. (2017). The role of the commensal microbiota in adaptive and maladaptive stressor-induced immunomodulation. Hormones & Behavior, 88, 7078. doi:10.1016/j.yhbeh.2016.10.006CrossRefGoogle ScholarPubMed
Maercker, A., Brewin, C. R., Bryant, R. A., Cloitre, M., van Ommeren, M., Jones, L. M., … Reed, G. M. (2013). Diagnosis and classification of disorders specifically associated with stress: Proposals for ICD-11. World Psychiatry, 12, 198206. doi:10.1002/wps.20057CrossRefGoogle ScholarPubMed
Maes, M., Lambrechts, J., Bosmans, E., Jacobs, J., Suy, E., Vandervorst, C., … Raus, J. (1992). Evidence for a systemic immune activation during depression: Results of leukocyte enumeration by flow cytometry in conjunction with monoclonal antibody staining. Psychological Medicine, 22, 4553. doi:10.1017/s0033291700032712CrossRefGoogle ScholarPubMed
Matthews, K. A., Chang, Y. F., Thurston, R. C., & Bromberger, J. T. (2014). Child abuse is related to inflammation in mid-life women: Role of obesity. Brain Behavior & Immunity, 36, 2934. doi:10.1016/j.bbi.2013.09.013CrossRefGoogle ScholarPubMed
Matthews, K., & Robbins, T. W. (2003). Early experience as a determinant of adult behavioural responses to reward: The effects of repeated maternal separation in the rat. Neuroscience & Biobehavioral Reviews, 27, 4555. doi:10.1016/s0149-7634(03)00008-3CrossRefGoogle ScholarPubMed
Matthews, K. A., Schott, L. L., Bromberger, J. T., Cyranowski, J. M., Everson-Rose, S. A., & Sowers, M. (2010). Are there bi-directional associations between depressive symptoms and C-reactive protein in mid-life women? Brain Behavior & Immunity, 24, 96101. doi:10.1016/j.bbi.2009.08.005CrossRefGoogle ScholarPubMed
McCrory, E. J., De Brito, S. A., Kelly, P. A., Bird, G., Sebastian, C. L., Mechelli, A., … Viding, E. (2013). Amygdala activation in maltreated children during pre-attentive emotional processing. British Journal of Psychiatry, 202, 269276. doi:10.1192/bjp.bp.112.116624CrossRefGoogle ScholarPubMed
McDade, T. W. (2012). Early environments and the ecology of inflammation. Proceedings of the National Academy of Sciences of the United States of America, 109, 1728117288. doi:10.1073/pnas.1202244109CrossRefGoogle ScholarPubMed
McDade, T. W., Borja, J. B., Adair, L. S., & Kuzawa, C. (2013a). Depressive symptoms are not associated with inflammation in younger and older adults in the Philippines. Evolution, Medicine, and Public Health, 2013, 1823. doi:10.1093/emph/eos004CrossRefGoogle Scholar
McDade, T. W., Burhop, J., & Dohnal, J. (2004). High-sensitivity enzyme immunoassay for C-reactive protein in dried blood spots. Clinical Chemistry, 50, 652654. doi:10.1373/clinchem.2003.029488CrossRefGoogle ScholarPubMed
McDade, T. W., Hoke, M., Borja, J. B., Adair, L. S., & Kuzawa, C. (2013b). Do environments in infancy moderate the association between stress and inflammation in adulthood? Initial evidence from a birth cohort in the Philippines. Brain Behavior & Immunity, 31, 2330. doi:10.1016/j.bbi.2012.08.010CrossRefGoogle Scholar
McDade, T. W., Rutherford, J., Adair, L., & Kuzawa, C. W. (2010). Early origins of inflammation: Microbial exposures in infancy predict lower levels of C-reactive protein in adulthood. Proceedings of the Royal Society B, 277, 11291137. doi:10.1098/rspb.2009.1795CrossRefGoogle Scholar
McDade, T. W., Tallman, P. S., Madimenos, F. C., Liebert, M. A., Cepon, T. J., Sugiyama, L. S., & Snodgrass, J. J. (2012). Analysis of variability of high sensitivity C-reactive protein in lowland Ecuador reveals no evidence of chronic low-grade inflammation. American Journal of Human Biology, 24, 675681. doi:10.1002/ajhb.22296CrossRefGoogle ScholarPubMed
McDade, T. W., Williams, S., & Snodgrass, J. J. (2007). What a drop can do: Dried blood spots as a minimally invasive method for integrating biomarkers into population-based research. Demography, 44, 899925. doi:10.1353/dem.2007.0038CrossRefGoogle Scholar
McEwen, B. S. (2007). Physiology and neurobiology of stress and adaptation: Central role of the brain. Physiological Reviews, 87, 873904. doi:10.1152/physrev.00041.2006CrossRefGoogle ScholarPubMed
McEwen, B. S. (2012). Brain on stress: How the social environment gets under the skin. Proceedings of the National Academy of Sciences of the United States of America, 109, 1718017185. doi:10.1073/pnas.1121254109CrossRefGoogle ScholarPubMed
McEwen, B. S., Weiss, J. M., & Schwartz, L. S. (1968). Selective retention of corticosterone by limbic structures in rat brain. Nature, 220, 911912. doi:10.1038/220911a0CrossRefGoogle ScholarPubMed
McIntyre, R. S., Subramaniapillai, M., Lee, Y., Pan, Z., Carmona, N. E., Shekotikhina, M., … Mansur, R. B. (2019). Efficacy of adjunctive infliximab vs placebo in the treatment of adults with Bipolar I/II depression: A randomized clinical trial. JAMA Psychiatry, 76, 783790. doi:10.1001/jamapsychiatry.2019.0779CrossRefGoogle ScholarPubMed
McLaughlin, K. A. (2016). Future directions in childhood adversity and youth psychopathology. Journal of Clinical Child and Adolescent Psychology, 45, 361382. doi:10.1080/15374416.2015.1110823CrossRefGoogle ScholarPubMed
McLaughlin, K. A., Greif Green, J., Gruber, M. J., Sampson, N. A., Zaslavsky, A. M., & Kessler, R. C. (2012). Childhood adversities and first onset of psychiatric disorders in a national sample of US adolescents. Archives of General Psychiatry, 69, 11511160. doi:10.1001/archgenpsychiatry.2011.2277CrossRefGoogle Scholar
McLaughlin, K. A., Sheridan, M. A., & Lambert, H. K. (2014). Childhood adversity and neural development: Deprivation and threat as distinct dimensions of early experience. Neuroscience & Biobehavioral Reviews, 47, 578591.CrossRefGoogle ScholarPubMed
McVey Neufeld, K. A., Mao, Y. K., Bienenstock, J., Foster, J. A., & Kunze, W. A. (2013). The microbiome is essential for normal gut intrinsic primary afferent neuron excitability in the mouse. Neurogastroenterology and Motility, 25, 183e188. doi:10.1111/nmo.12049CrossRefGoogle ScholarPubMed
Measelle, J. R., & Ablow, J. C. (2018). Contributions of early adversity to pro-inflammatory phenotype in infancy: The buffer provided by attachment security. Attachment and Human Development, 20, 123. doi:10.1080/14616734.2017.1362657CrossRefGoogle ScholarPubMed
Mednick, S. A., Machon, R. A., Huttunen, M. O., & Bonett, D. (1988). Adult schizophrenia following prenatal exposure to an influenza epidemic. Archives of General Psychiatry, 45, 189192. doi:10.1001/archpsyc.1988.01800260109013CrossRefGoogle Scholar
Medzhitov, R. (2008). Origin and physiological roles of inflammation. Nature, 454, 428435. doi:10.1038/nature07201CrossRefGoogle ScholarPubMed
Mehta, M. A., Gore-Langton, E., Golembo, N., Colvert, E., Williams, S. C., & Sonuga-Barke, E. (2010). Hyporesponsive reward anticipation in the basal ganglia following severe institutional deprivation early in life. Journal of Cognitive Neuroscience, 22, 23162325. doi:10.1162/jocn.2009.21394CrossRefGoogle ScholarPubMed
Michels, N., Sioen, I., Braet, C., Eiben, G., Hebestreit, A., Huybrechts, I., … De Henauw, S. (2012). Stress, emotional eating behaviour and dietary patterns in children. Appetite, 59, 762769. doi:10.1016/j.appet.2012.08.010CrossRefGoogle ScholarPubMed
Michigan, A., Johnson, T. V., & Master, V. A. (2011). Review of the relationship between C-reactive protein and exercise. Molecular Diagnosis & Therapy, 15, 265275. doi:10.1007/bf03256418CrossRefGoogle ScholarPubMed
Michopoulos, V., Rothbaum, A. O., Jovanovic, T., Almli, L. M., Bradley, B., Rothbaum, B. O., … Ressler, K. J. (2015). Association of CRP genetic variation and CRP level with elevated PTSD symptoms and physiological responses in a civilian population with high levels of trauma. American Journal of Psychiatry, 172, 353362. doi:10.1176/appi.ajp.2014.14020263CrossRefGoogle Scholar
Miller, B. J., Buckley, P., Seabolt, W., Mellor, A., & Kirkpatrick, B. (2011a). Meta-analysis of cytokine alterations in schizophrenia: Clinical status and antipsychotic effects. Biological Psychiatry, 70, 663671. doi:10.1016/j.biopsych.2011.04.013CrossRefGoogle Scholar
Miller, G. E., & Chen, E. (2010). Harsh family climate in early life presages the emergence of a proinflammatory phenotype in adolescence. Psychological Sciences, 21, 848856. doi:10.1177/0956797610370161CrossRefGoogle ScholarPubMed
Miller, G. E., Chen, E., & Parker, K. J. (2011b). Psychological stress in childhood and susceptibility to the chronic diseases of aging: Moving toward a model of behavioral and biological mechanisms. Psychological Bulletin, 137, 959997. doi:10.1037/a0024768CrossRefGoogle Scholar
Miller, G. E., Cohen, S., & Ritchey, A. K. (2002). Chronic psychological stress and the regulation of pro-inflammatory cytokines: A glucocorticoid-resistance model. Health Psychology, 21, 531541. doi:10.1037//0278-6133.21.6.531CrossRefGoogle ScholarPubMed
Miller, G. E., & Cole, S. W. (2012). Clustering of depression and inflammation in adolescents previously exposed to childhood adversity. Biological Psychiatry, 72, 3440. doi:10.1016/j.biopsych.2012.02.034CrossRefGoogle ScholarPubMed
Miller, G. E., Freedland, K. E., Carney, R. M., Stetler, C. A., & Banks, W. A. (2003). Pathways linking depression, adiposity, and inflammatory markers in healthy young adults. Brain Behavior & Immunity, 17, 276285. doi:10.1016/s0889-1591(03)00057-6CrossRefGoogle ScholarPubMed
Miller, M. A., Kandala, N.-B., Kivimaki, M., Kumari, M., Brunner, E. J., Lowe, G., … Cappuccio, F. P. (2009). Gender differences in the cross-sectional relationships between sleep duration and markers of inflammation: Whitehall II study. Sleep, 32, 857864. doi:10.1093/sleep/32.7.857Google Scholar
Miller, A. H., & Raison, C. L. (2015). The role of inflammation in depression: From evolutionary imperative to modern treatment target. Nature Reviews Immunology, 16(1), 2234CrossRefGoogle Scholar
Miller, A. H., & Raison, C. L. (2016). The role of inflammation in depression: From evolutionary imperative to modern treatment target. Nature Reviews Immunology, 16, 2234. doi:10.1038/nri.2015.5CrossRefGoogle Scholar
Modabbernia, A., Taslimi, S., Brietzke, E., & Ashrafi, M. (2013). Cytokine alterations in bipolar disorder: A meta-analysis of 30 studies. Biological Psychiatry, 74, 1525. doi:10.1016/j.biopsych.2013.01.007CrossRefGoogle ScholarPubMed
Mondelli, V., Ciufolini, S., Belvederi Murri, M., Bonaccorso, S., Di Forti, M., Giordano, A., … Dazzan, P. (2015). Cortisol and inflammatory biomarkers predict poor treatment response in first episode psychosis. Schizophrenia Bulletin, 41, 11621170. doi:10.1093/schbul/sbv028CrossRefGoogle ScholarPubMed
Monje, M. L., Toda, H., & Palmer, T. D. (2003). Inflammatory blockade restores adult hippocampal neurogenesis. Science, 302, 17601765. doi:10.1126/science.1088417CrossRefGoogle ScholarPubMed
Monk, C., Georgieff, M. K., & Osterholm, E. A. (2013). Research review: Maternal prenatal distress and poor nutrition - mutually influencing risk factors affecting infant neurocognitive development. Journal of Child Psychology and Psychiatry, 54, 115130. doi:10.1111/jcpp.12000CrossRefGoogle ScholarPubMed
Moreira, A. P., Texeira, T. F., Ferreira, A. B., Peluzio Mdo, C., & Alfenas Rde, C. (2012). Influence of a high-fat diet on gut microbiota, intestinal permeability and metabolic endotoxaemia. British Journal of Nutrition, 108, 801809. doi:10.1017/S0007114512001213CrossRefGoogle ScholarPubMed
Mrdalj, J., Pallesen, S., Milde, A. M., Jellestad, F. K., Murison, R., Ursin, R., … Gronli, J. (2013). Early and later life stress alter brain activity and sleep in rats. PLoS One, 8, e69923. doi:10.1371/journal.pone.0069923CrossRefGoogle ScholarPubMed
Murphy, K. (2011). Janeway's immunobiology. New York: Garland Sci.Google Scholar
Musselman, D. L., Lawson, D. H., Gumnick, J. F., Manatunga, A. K., Penna, S., Goodkin, R. S., … Miller, A. H. (2001). Paroxetine for the prevention of depression induced by high-dose interferon alfa. The New England Journal of Medicine, 344, 961966. doi:10.1056/NEJM200103293441303CrossRefGoogle ScholarPubMed
Nanni, V., Uher, R., & Danese, A. (2012). Childhood maltreatment predicts unfavorable course of illness and treatment outcome in depression: A meta-analysis. American Journal of Psychiatry, 169, 141151. doi:10.1176/appi.ajp.2011.11020335CrossRefGoogle ScholarPubMed
Nanri, A., Kimura, Y., Matsushita, Y., Ohta, M., Sato, M., Mishima, N., … Mizoue, T. (2010). Dietary patterns and depressive symptoms among Japanese men and women. European Journal of Clinical Nutrition, 64, 832. doi:10.1038/ejcn.2010.86CrossRefGoogle ScholarPubMed
Network, & Pathway Analysis Subgroup of Psychiatric Genomics, C. (2015). Psychiatric genome-wide association study analyses implicate neuronal, immune and histone pathways. Nature Neuroscience, 18, 199209. doi:10.1038/nn.3922CrossRefGoogle Scholar
Noll, J. G., Trickett, P. K., Susman, E. J., & Putnam, F. W. (2006). Sleep disturbances and childhood sexual abuse. Journal of Pediatric Psychology, 31, 469480. doi:10.1093/jpepsy/jsj040CrossRefGoogle ScholarPubMed
Nusslock, R., & Miller, G. E. (2016). Early-life adversity and physical and emotional health across the lifespan: A neuroimmune network hypothesis. Biological Psychiatry, 80, 2332. doi:10.1016/j.biopsych.2015.05.017CrossRefGoogle ScholarPubMed
O'Connor, T. G., Willoughby, M. T., Moynihan, J. A., Messing, S., Vallejo Sefair, A., Carnahan, J., … Caserta, M. T. (2020). Early childhood risk exposures and inflammation in early adolescence. Brain Behavior & Immunity, 86, 2229. doi: 10.1016/j.bbi.2019.05.001CrossRefGoogle ScholarPubMed
O'Mahony, S. M., Marchesi, J. R., Scully, P., Codling, C., Ceolho, A. M., Quigley, E. M., … Dinan, T. G. (2009). Early life stress alters behavior, immunity, and microbiota in rats: Implications for irritable bowel syndrome and psychiatric illnesses. Biological Psychiatry, 65, 263267. doi:10.1016/j.biopsych.2008.06.026CrossRefGoogle ScholarPubMed
Osborn, M., & Widom, C. S. (2019). Do documented records and retrospective reports of childhood maltreatment similarly predict chronic inflammation? Psychological Medicine, 110. doi:10.1017/S0033291719002575Google ScholarPubMed
Ouellet-Morin, I., Danese, A., Williams, B., & Arseneault, L. (2011). Validation of a high-sensitivity assay for C-reactive protein in human saliva. Brain Behavior & Immunity, 25, 640646. doi:10.1016/j.bbi.2010.12.020CrossRefGoogle ScholarPubMed
Pace, T. W., Mletzko, T. C., Alagbe, O., Musselman, D. L., Nemeroff, C. B., Miller, A. H., & Heim, C. M. (2006). Increased stress-induced inflammatory responses in male patients with major depression and increased early life stress. American Journal of Psychiatry, 163, 16301633. doi:10.1176/ajp.2006.163.9.1630CrossRefGoogle ScholarPubMed
Panagiotaropoulos, T., Papaioannou, A., Pondiki, S., Prokopiou, A., Stylianopoulou, F., & Gerozissis, K. (2004). Effect of neonatal handling and sex on basal and chronic stress-induced corticosterone and leptin secretion. Neuroendocrinology, 79, 109118. doi:10.1159/000076633CrossRefGoogle ScholarPubMed
Pankow, J. S., Folsom, A. R., Cushman, M., Borecki, I. B., Hopkins, P. N., Eckfeldt, J. H., & Tracy, R. P. (2001). Familial and genetic determinants of systemic markers of inflammation: The NHLBI family heart study. Atherosclerosis, 154, 681689. doi:10.1016/s0021-9150(00)00586-4CrossRefGoogle Scholar
Paolicelli, R. C., Bolasco, G., Pagani, F., Maggi, L., Scianni, M., Panzanelli, P., … Gross, C. T. (2011). Synaptic pruning by microglia is necessary for normal brain development. Science, 333, 14561458. doi:10.1126/science.1202529CrossRefGoogle ScholarPubMed
Pariante, C. M., & Miller, A. H. (2001). Glucocorticoid receptors in major depression: Relevance to pathophysiology and treatment. Biological Psychiatry, 49, 391404. doi:10.1016/s0006-3223(00)01088-xCrossRefGoogle ScholarPubMed
Parikh, S., Pollock, N. K., Bhagatwala, J., Guo, D.-H., Gutin, B., Zhu, H., & Dong, Y. (2012). Adolescent fiber consumption is associated with visceral fat and inflammatory markers. The Journal of Clinical Endocrinology & Metabolism, 97, E1451E1457. doi:10.1210/jc.2012-1784CrossRefGoogle ScholarPubMed
Park, H., Tsai, K. M., Dahl, R. E., Irwin, M. R., McCreath, H., Seeman, T. E., & Fuligni, A. J. (2016). Sleep and inflammation during adolescence. Psychosomatic Medicine, 78, 677685. doi:10.1097/PSY.0000000000000340CrossRefGoogle ScholarPubMed
Pasco, J. A., Williams, L. J., Jacka, F. N., Henry, M. J., Coulson, C. E., Brennan, S. L., … Berk, M. (2011). Habitual physical activity and the risk for depressive and anxiety disorders among older men and women. International Psychogeriatrics, 23, 292298. doi:10.1017/s1041610210001833CrossRefGoogle ScholarPubMed
Patterson, P. H. (2009). Immune involvement in schizophrenia and autism: Etiology, pathology and animal models. Behavioral Brain Research, 204, 313321. doi:10.1016/j.bbr.2008.12.016CrossRefGoogle ScholarPubMed
Pechtel, P., & Pizzagalli, D. A. (2011). Effects of early life stress on cognitive and affective function: An integrated review of human literature. Psychopharmacology (Berl), 214, 5570. doi:10.1007/s00213-010-2009-2CrossRefGoogle ScholarPubMed
Peckins, M. K., Dockray, S., Eckenrode, J. L., Heaton, J., & Susman, E. J. (2012). The longitudinal impact of exposure to violence on cortisol reactivity in adolescents. Journal of Adolescent Health, 51, 366372. doi:10.1016/j.jadohealth.2012.01.005CrossRefGoogle ScholarPubMed
Peferoen, L., Kipp, M., van der Valk, P., van Noort, J. M., & Amor, S. (2014). Oligodendrocyte-microglia cross-talk in the central nervous system. Immunology, 141, 302313. doi:10.1111/imm.12163CrossRefGoogle ScholarPubMed
Perry, V. H., & Holmes, C. (2014). Microglial priming in neurodegenerative disease. Nature Reviews Neurology, 10, 217224. doi:10.1038/nrneurol.2014.38CrossRefGoogle ScholarPubMed
Phillips, G. D., Howes, S. R., Whitelaw, R. B., Robbins, T. W., & Everitt, B. J. (1994). Isolation rearing impairs the reinforcing efficacy of intravenous cocaine or intra-accumbens d-amphetamine: Impaired response to intra-accumbens D1 and D2/D3 dopamine receptor antagonists. Psychopharmacology (Berl), 115, 419429. doi:10.1007/bf02245085CrossRefGoogle ScholarPubMed
Pinto Pereira, S. M., Stein Merkin, S., Seeman, T., & Power, C. (2019). Understanding associations of early-life adversities with mid-life inflammatory profiles: Evidence from the UK and USA. Brain Behavior & Immunity, 78, 143152. doi:10.1016/j.bbi.2019.01.016CrossRefGoogle ScholarPubMed
Pistell, P. J., Morrison, C. D., Gupta, S., Knight, A. G., Keller, J. N., Ingram, D. K., & Bruce-Keller, A. J. (2010). Cognitive impairment following high fat diet consumption is associated with brain inflammation. Journal of Neuroimmunology, 219, 2532. doi:10.1016/j.jneuroim.2009.11.010CrossRefGoogle ScholarPubMed
Pollak, S. D., & Kistler, D. J. (2002). Early experience is associated with the development of categorical representations for facial expressions of emotion. Proceedings of the National Academy of Sciences of the United States of America, 99, 90729076. doi:10.1073/pnas.142165999CrossRefGoogle ScholarPubMed
Prather, A. A., Hall, M., Fury, J. M., Ross, D. C., Muldoon, M. F., Cohen, S., & Marsland, A. L. (2012). Sleep and antibody response to hepatitis B vaccination. Sleep, 35, 10631069. doi:10.5665/sleep.1990Google ScholarPubMed
Pryce, C. R., Dettling, A., Spengler, M., Spaete, C., & Feldon, J. (2004). Evidence for altered monoamine activity and emotional and cognitive disturbance in marmoset monkeys exposed to early life stress. Annals of the New York Academy of Science, 1032, 245249. doi:10.1196/annals.1314.030CrossRefGoogle ScholarPubMed
Raison, C. L., Rutherford, R. E., Woolwine, B. J., Shuo, C., Schettler, P., Drake, D. F., … Miller, A. H. (2013). A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: The role of baseline inflammatory biomarkers. JAMA Psychiatry, 70, 3141. doi:10.1001/2013.jamapsychiatry.4CrossRefGoogle ScholarPubMed
Rangel-Huerta, O. D., Aguilera, C. M., Mesa, M. D., & Gil, A. (2012). Omega-3 long-chain polyunsaturated fatty acids supplementation on inflammatory biomakers: A systematic review of randomised clinical trials. British Journal of Nutrition, 107, S159170. doi:10.1017/S0007114512001559CrossRefGoogle ScholarPubMed
Raposa, E. B., Bower, J. E., Hammen, C. L., Najman, J. M., & Brennan, P. A. (2014). A developmental pathway from early life stress to inflammation: The role of negative health behaviors. Psychological Science, 25, 12681274. doi:10.1177/0956797614530570CrossRefGoogle ScholarPubMed
Reichenberg, A., Yirmiya, R., Schuld, A., Kraus, T., Haack, M., Morag, A., & Pollmacher, T. (2001). Cytokine-associated emotional and cognitive disturbances in humans. Archives of General Psychiatry, 58, 445452. doi:10.1001/archpsyc.58.5.445CrossRefGoogle ScholarPubMed
Reid, B. M., Coe, C. L., Doyle, C. M., Sheerar, D., Slukvina, A., Donzella, B., & Gunnar, M. R. (2019). Persistent skewing of the T-cell profile in adolescents adopted internationally from institutional care. Brain Behavior & Immunity, 77, 168177. doi:10.1016/j.bbi.2019.01.001CrossRefGoogle ScholarPubMed
Reid, B. M., Doom, J. R., Argote, R. B., Correa-Burrows, P., Lozoff, B., Blanco, E., & Gahagan, S. (2020). Pathways to inflammation in adolescence through early adversity, childhood depressive symptoms, and body mass index: A prospective longitudinal study of Chilean infants. Brain Behavior & Immunity, 86, 413. doi: 10.1016/j.bbi.2019.06.003CrossRefGoogle ScholarPubMed
Reuben, A., Moffitt, T. E., Caspi, A., Belsky, D. W., Harrington, H., Schroeder, F., … Danese, A. (2016). Lest we forget: Comparing retrospective and prospective assessments of adverse childhood experiences in the prediction of adult health. Journal of Child Psychology and Psychiatry, 57, 11031112. doi:10.1111/jcpp.12621CrossRefGoogle ScholarPubMed
Riis, J. L., Granger, D. A., Minkovitz, C. S., Bandeen-Roche, K., DiPietro, J. A., & Johnson, S. B. (2016). Maternal distress and child neuroendocrine and immune regulation. Social Science & Medicine, 151, 206214. doi:10.1016/j.socscimed.2015.12.043CrossRefGoogle ScholarPubMed
Rohleder, N., Nater, U. M., Wolf, J. M., Ehlert, U., & Kirschbaum, C. (2004). Psychosocial stress-induced activation of salivary alpha-amylase. Annals of the New York Academy of Sciences, 1032, 258263.CrossRefGoogle ScholarPubMed
Rooks, C., Veledar, E., Goldberg, J., Bremner, J. D., & Vaccarino, V. (2012). Early trauma and inflammation: Role of familial factors in a study of twins. Psychosomatic Medicine, 74, 146152. doi:10.1097/PSY.0b013e318240a7d8CrossRefGoogle Scholar
Runsten, S., Korkeila, K., Koskenvuo, M., Rautava, P., Vainio, O., & Korkeila, J. (2014). Can social support alleviate inflammation associated with childhood adversities? Nordic Journal of Psychiatry, 68, 137144. doi:10.3109/08039488.2013.786133CrossRefGoogle ScholarPubMed
Sanchez, M. M., Ladd, C. O., & Plotsky, P. M. (2001). Early adverse experience as a developmental risk factor for later psychopathology: Evidence from rodent and primate models. Development & Psychopathology, 13, 419450.CrossRefGoogle ScholarPubMed
Schaefer, J. D., Moffitt, T. E., Arseneault, L., Danese, A., Fisher, H. L., Houts, R., … Caspi, A. (2018). Adolescent victimization and early-adult psychopathology: Approaching causal inference using a longitudinal twin study to rule out noncausal explanations. Clinical Psychological Science, 6, 352371. doi:10.1177/2167702617741381CrossRefGoogle ScholarPubMed
Schedlowski, M., Engler, H., & Grigoleit, J. S. (2014). Endotoxin-induced experimental systemic inflammation in humans: A model to disentangle immune-to-brain communication. Brain Behavior & Immunity, 35, 18. doi:10.1016/j.bbi.2013.09.015CrossRefGoogle Scholar
Scrivo, R., Vasile, M., Bartosiewicz, I., & Valesini, G. (2011). Inflammation as “common soil” of the multifactorial diseases. Autoimmunity Reviews, 10(7), 369374.CrossRefGoogle ScholarPubMed
Segerstrom, S. C., & Miller, G. E. (2004). Psychological stress and the human immune system: A meta-analytic study of 30 years of inquiry. Psychological Bulletin, 130, 601630. doi:10.1037/0033-2909.130.4.601CrossRefGoogle ScholarPubMed
Shanks, N., Larocque, S., & Meaney, M. J. (1995). Neonatal endotoxin exposure alters the development of the hypothalamic-pituitary-adrenal axis: Early illness and later responsivity to stress. Journal of Neuroscience, 15, 376384. doi:10.1523/jneurosci.15-01-00376.1995CrossRefGoogle ScholarPubMed
Shanks, N., & Lightman, S. L. (2001). The maternal-neonatal neuro-immune interface: Are there long-term implications for inflammatory or stress-related disease? Journal of Clinical Investigation, 108, 15671573. doi:10.1172/JCI14592CrossRefGoogle ScholarPubMed
Shanks, N., Windle, R. J., Perks, P. A., Harbuz, M. S., Jessop, D. S., Ingram, C. D., & Lightman, S. L. (2000). Early-life exposure to endotoxin alters hypothalamic-pituitary-adrenal function and predisposition to inflammation. Proceedings of the National Academy of Sciences of the United States of America, 97, 56455650. doi:10.1073/pnas.090571897CrossRefGoogle ScholarPubMed
Shatz, C. J. (2009). MHC class I: An unexpected role in neuronal plasticity. Neuron, 64, 4045. doi:10.1016/j.neuron.2009.09.044CrossRefGoogle ScholarPubMed
Shields, G. S., Slavich, G. M., Perlman, G., Klein, D. N., & Kotov, R. (2019). The short-term reliability and long-term stability of salivary immune markers. Brain Behavior & Immunity, 81, 650654. doi:10.1016/j.bbi.2019.06.007CrossRefGoogle ScholarPubMed
Shiels, M. S., Katki, H. A., Freedman, N. D., Purdue, M. P., Wentzensen, N., Trabert, B., … Chaturvedi, A. K. (2014). Cigarette smoking and variations in systemic immune and inflammation markers. Journal of the National Cancer Institute, 106, 11. doi:10.1093/jnci/dju294CrossRefGoogle ScholarPubMed
Shirtcliff, E. A., Coe, C. L., & Pollak, S. D. (2009). Early childhood stress is associated with elevated antibody levels to herpes simplex virus type 1. Proceedings of the National Academy of Sciences of the United States of America, 106, 29632967. doi:10.1073/pnas.0806660106CrossRefGoogle ScholarPubMed
Short, S. J., Lubach, G. R., Karasin, A. I., Olsen, C. W., Styner, M., Knickmeyer, R. C., … Coe, C. L. (2010). Maternal influenza infection during pregnancy impacts postnatal brain development in the rhesus monkey. Biological Psychiatry, 67, 965973. doi:10.1016/j.biopsych.2009.11.026CrossRefGoogle ScholarPubMed
Slavich, G. M., & Irwin, M. R. (2014). From stress to inflammation and major depressive disorder: A social signal transduction theory of depression. Psychological Bulletin, 140, 774815. doi:10.1037/a0035302CrossRefGoogle ScholarPubMed
Slavish, D. C., Graham-Engeland, J. E., Smyth, J. M., & Engeland, C. G. (2015). Salivary markers of inflammation in response to acute stress. Brain Behavior & Immunity, 44, 253269. doi:10.1016/j.bbi.2014.08.008CrossRefGoogle Scholar
Slopen, N., Lewis, T. T., Gruenewald, T. L., Mujahid, M. S., Ryff, C. D., Albert, M. A., & Williams, D. R. (2010). Early life adversity and inflammation in African Americans and whites in the midlife in the United States survey. Psychosomatic Medicine, 72, 694701. doi:10.1097/PSY.0b013e3181e9c16fCrossRefGoogle ScholarPubMed
Slopen, N., Tang, A., Nelson, C. A., Zeanah, C. H., McDade, T. W., McLaughlin, K. A., & Fox, N. A. (2019). The consequences of foster care versus institutional care in early childhood on adolescent cardiometabolic and immune markers: Results from a randomized controlled trial. Psychosomatic Medicine, 81, 449457. doi:10.1097/PSY.0000000000000696CrossRefGoogle ScholarPubMed
Smith, R. S. (1991). The macrophage theory of depression. Medical Hypotheses, 35, 298306. doi:10.1016/0306-9877(91)90272-zCrossRefGoogle ScholarPubMed
Smith, A. K., Conneely, K. N., Kilaru, V., Mercer, K. B., Weiss, T. E., Bradley, B., … Ressler, K. J. (2011). Differential immune system DNA methylation and cytokine regulation in post-traumatic stress disorder. American Journal of Medical Genetics Part B-Neuropsychiatric Genetics, 156B, 700708. doi:10.1002/ajmg.b.31212CrossRefGoogle ScholarPubMed
Spiegel, K., Sheridan, J. F., & Van Cauter, E. (2002). Effect of sleep deprivation on response to immunization. JAMA, 288, 14711472. doi:10.1001/jama.288.12.1471-aCrossRefGoogle ScholarPubMed
Stellwagen, D., & Malenka, R. C. (2006). Synaptic scaling mediated by glial TNF-alpha. Nature, 440, 10541059. doi:10.1038/nature04671CrossRefGoogle ScholarPubMed
Stephan, A. H., Barres, B. A., & Stevens, B. (2012). The complement system: an unexpected role in synaptic pruning during development and disease. Annual Review of Neuroscience, 35, 369389.CrossRefGoogle ScholarPubMed
Stevens, G. A., Finucane, M. M., De-Regil, L. M., Paciorek, C. J., Flaxman, S. R., Branca, F., … Nutrition Impact Model Study, G. (2013). Global, regional, and national trends in haemoglobin concentration and prevalence of total and severe anaemia in children and pregnant and non-pregnant women for 1995-2011: A systematic analysis of population-representative data. Lancet Global Health, 1, e1625. doi:10.1016/S2214-109X(13)70001-9CrossRefGoogle ScholarPubMed
Strawbridge, R., Arnone, D., Danese, A., Papadopoulos, A., Herane Vives, A., & Cleare, A. J. (2015). Inflammation and clinical response to treatment in depression: A meta-analysis. Eur Neuropsychopharmacology, 25, 15321543. doi:10.1016/j.euroneuro.2015.06.007CrossRefGoogle ScholarPubMed
Strawbridge, W. J., Deleger, S., Roberts, R. E., & Kaplan, G. A. (2002). Physical activity reduces the risk of subsequent depression for older adults. American Journal of Epidemiology, 156, 328334. doi:10.1093/aje/kwf047CrossRefGoogle ScholarPubMed
Suchdev, P. S., Boivin, M. J., Forsyth, B. W., Georgieff, M. K., Guerrant, R. L., & Nelson, C. A. 3rd. (2017). Assessment of neurodevelopment, nutrition, and inflammation from fetal life to adolescence in low-resource settings. Pediatrics, 139, S23S37. doi:10.1542/peds.2016-2828ECrossRefGoogle ScholarPubMed
Suomi, S. J. (1997). Early determinants of behaviour: Evidence from primate studies. Br Med Bull, 53, 170184. doi:10.1093/oxfordjournals.bmb.a011598CrossRefGoogle ScholarPubMed
Tachon, S., Zhou, J., Keenan, M., Martin, R., & Marco, M. L. (2013). The intestinal microbiota in aged mice is modulated by dietary resistant starch and correlated with improvements in host responses. FEMS Microbiology Ecology, 83, 299309. doi:10.1111/j.1574-6941.2012.01475.xCrossRefGoogle ScholarPubMed
Takatsuru, Y., Nabekura, J., Ishikawa, T., Kohsaka, S., & Koibuchi, N. (2015). Early-life stress increases the motility of microglia in adulthood. Journal of Physiological Science, 65, 187194. doi:10.1007/s12576-015-0361-zCrossRefGoogle ScholarPubMed
Tang, A., Slopen, N., Nelson, C. A., Zeanah, C. H., Georgieff, M. K., & Fox, N. A. (2018). Catch-up growth, metabolic, and cardiovascular risk in post-institutionalized Romanian adolescents. Pediatric Research, 84, 842848. doi:10.1038/s41390-018-0196-4CrossRefGoogle ScholarPubMed
Tarullo, A. R., & Gunnar, M. R. (2006). Child maltreatment and the developing HPA axis. Hormones & Behavior, 50, 632639. doi:10.1016/j.yhbeh.2006.06.010CrossRefGoogle ScholarPubMed
Taylor, S. E., Lehman, B. J., Kiefe, C. I., & Seeman, T. E. (2006). Relationship of early life stress and psychological functioning to adult C-reactive protein in the coronary artery risk development in young adults study. Biological Psychiatry, 60, 819824. doi:10.1016/j.biopsych.2006.03.016CrossRefGoogle ScholarPubMed
Teicher, M. H., & Samson, J. A. (2013). Childhood maltreatment and psychopathology: A case for ecophenotypic variants as clinically and neurobiologically distinct subtypes. American Journal of Psychiatry, 170, 11141133. doi:10.1176/appi.ajp.2013.12070957CrossRefGoogle ScholarPubMed
Toth, L. A. (1995). Sleep, sleep deprivation and infectious disease: Studies in animals. Advances in Neuroimmunology, 5, 7992. doi:10.1016/0960-5428(94)00045-pCrossRefGoogle ScholarPubMed
Tottenham, N., Hare, T. A., Millner, A., Gilhooly, T., Zevin, J. D., & Casey, B. J. (2011). Elevated amygdala response to faces following early deprivation. Developmental Science, 14, 190204. doi:10.1111/j.1467-7687.2010.00971.xCrossRefGoogle ScholarPubMed
Tran, P. B., & Miller, R. J. (2003). Chemokine receptors: Signposts to brain development and disease. Nature Review Neuroscience, 4, 444455. doi:10.1038/nrn1116CrossRefGoogle ScholarPubMed
Tran, P., Singh, G., Wallin, D., & Georgieff, M. (2018). Neonatal phlebotomy-induced anemia in developing female mice increases immune response genes and decreases estrogen signaling in the hippocampus. Paper presented at the Pediatric Academic Societies Meeting, Toronto, Canada.Google Scholar
Tyrka, A. R., Parade, S. H., Valentine, T. R., Eslinger, N. M., & Seifer, R. (2015). Adversity in preschool-aged children: Effects on salivary interleukin-1beta. Development and Psychopathology, 27, 567576. doi:10.1017/S0954579415000164CrossRefGoogle ScholarPubMed
Valkanova, V., Ebmeier, K. P., & Allan, C. L. (2013). CRP, IL-6 and depression: A systematic review and meta-analysis of longitudinal studies. Journal of Affective Disorders, 150, 736744. doi:10.1016/j.jad.2013.06.004CrossRefGoogle ScholarPubMed
Varese, F., Smeets, F., Drukker, M., Lieverse, R., Lataster, T., Viechtbauer, W., … Bentall, R. P. (2012). Childhood adversities increase the risk of psychosis: A meta-analysis of patient-control, prospective- and cross-sectional cohort studies. Schizophrenia Bulletin, 38, 661671. doi:10.1093/schbul/sbs050CrossRefGoogle ScholarPubMed
Visser, M., Bouter, L. M., McQuillan, G. M., Wener, M. H., & Harris, T. B. (2001). Low-grade systemic inflammation in overweight children. Pediatrics, 107, E13. doi:10.1542/peds.107.1.e13CrossRefGoogle ScholarPubMed
Viviani, B., Boraso, M., Valero, M., Gardoni, F., Marco, E. M., Llorente, R., … Viveros, M. P. (2014). Early maternal deprivation immunologically primes hippocampal synapses by redistributing interleukin-1 receptor type I in a sex dependent manner. Brain Behavior & Immunity, 35, 135143. doi:10.1016/j.bbi.2013.09.008CrossRefGoogle Scholar
Volman, J. J., Ramakers, J. D., & Plat, J. (2008). Dietary modulation of immune function by beta-glucans. Physiology & Behavior, 94, 276284. doi:10.1016/j.physbeh.2007.11.045CrossRefGoogle ScholarPubMed
Walsh, K., Basu, A., Werner, E., Lee, S., Feng, T., Osborne, L. M., … Monk, C. (2016). Associations among child abuse, depression, and interleukin-6 in pregnant adolescents: Paradoxical findings. Psychosomatic Medicine, 78, 920930. doi:10.1097/PSY.0000000000000344CrossRefGoogle ScholarPubMed
Watzl, B., Kulling, S. E., Moseneder, J., Barth, S. W., & Bub, A. (2005). A 4-wk intervention with high intake of carotenoid-rich vegetables and fruit reduces plasma C-reactive protein in healthy, nonsmoking men. American Journal of Clinical Nutrition, 82, 10521058. doi:10.1093/ajcn/82.5.1052CrossRefGoogle ScholarPubMed
Weaver, I. C., Cervoni, N., Champagne, F. A., D'Alessio, A. C., Sharma, S., Seckl, J. R., … Meaney, M. J. (2004). Epigenetic programming by maternal behavior. Nature Neuroscience, 7, 847854. doi:10.1038/nn1276CrossRefGoogle ScholarPubMed
Wei, L., Simen, A., Mane, S., & Kaffman, A. (2012). Early life stress inhibits expression of a novel innate immune pathway in the developing hippocampus. Neuropsychopharmacology, 37, 567580. doi:10.1038/npp.2011.239CrossRefGoogle ScholarPubMed
Weinberg, R. J., Dietz, L. J., Stoyak, S., Melhem, N. M., Porta, G., Payne, M. W., & Brent, D. A. (2013). A prospective study of parentally bereaved youth, caregiver depression, and body mass index. Journal of Clinical Psychiatry, 74, 834840. doi:10.4088/JCP.12m08284CrossRefGoogle Scholar
Wells, J. M., Brummer, R. J., Derrien, M., MacDonald, T. T., Troost, F., Cani, P. D., … Garcia-Rodenas, C. L. (2017). Homeostasis of the gut barrier and potential biomarkers. American Journal of Physiology Gastrointestinal Liver Physiology, 312, G171G193. doi:10.1152/ajpgi.00048.2015CrossRefGoogle ScholarPubMed
Weng, T.-T., Hao, J.-H., Qian, Q.-W., Cao, H., Fu, J.-L., Sun, Y., … Tao, F.-B. (2012). Is there any relationship between dietary patterns and depression and anxiety in Chinese adolescents? Public Health Nutrition, 15, 673682. doi:10.1017/s1368980011003077CrossRefGoogle ScholarPubMed
Widom, C. S. (1999). Posttraumatic stress disorder in abused and neglected children grown up. American Journal of Psychiatry, 156, 12231229. doi:10.1176/ajp.156.8.1223Google ScholarPubMed
Wieck, A., Andersen, S. L., & Brenhouse, H. C. (2013). Evidence for a neuroinflammatory mechanism in delayed effects of early life adversity in rats: Relationship to cortical NMDA receptor expression. Brain Behavior & Immunity, 28, 218226. doi:10.1016/j.bbi.2012.11.012CrossRefGoogle ScholarPubMed
Yirmiya, R., & Goshen, I. (2011). Immune modulation of learning, memory, neural plasticity and neurogenesis. Brain Behavior & Immunity, 25, 181213. doi:10.1016/j.bbi.2010.10.015CrossRefGoogle ScholarPubMed
Yuan, N., Chen, Y., Xia, Y., Dai, J., & Liu, C. (2019). Inflammation-related biomarkers in major psychiatric disorders: A cross-disorder assessment of reproducibility and specificity in 43 meta-analyses. Translational Psychiatry, 9, 233. doi:10.1038/s41398-019-0570-yCrossRefGoogle ScholarPubMed
Zanella Terrier, M. C., Simonet, M. L., Bichard, P., & Frossard, J. L. (2014). Recurrent Clostridium difficile infections: The importance of the intestinal microbiota. World Journal of Gastroenterology, 20, 74167423. doi:10.3748/wjg.v20.i23.7416CrossRefGoogle ScholarPubMed
Zautra, A. J., Davis, M. C., Reich, J. W., Nicassario, P., Tennen, H., Finan, P., … Irwin, M. R. (2008). Comparison of cognitive behavioral and mindfulness meditation interventions on adaptation to rheumatoid arthritis for patients with and without history of recurrent depression. Journal of Consult Clinical Psychology, 76, 408421. doi:10.1037/0022-006X.76.3.408CrossRefGoogle ScholarPubMed
Zeugmann, S., Buehrsch, N., Bajbouj, M., Heuser, I., Anghelescu, I., & Quante, A. (2013). Childhood maltreatment and adult proinflammatory status in patients with major depression. Psychiatria Danubina, 25, 227235. Retrieved from <Go to ISI>://WOS:000338630000005.Google ScholarPubMed
Ziv, Y., Ron, N., Butovsky, O., Landa, G., Sudai, E., Greenberg, N., … Schwartz, M. (2006). Immune cells contribute to the maintenance of neurogenesis and spatial learning abilities in adulthood. Nature Neuroscience, 9, 268275. doi:10.1038/nn1629CrossRefGoogle Scholar
Figure 0

Table 1. Studies of early life adversity and peripheral inflammation, arranged by mean age (years) at inflammation assessment

Figure 1

Table 2. Early adversity and inflammation: adult at inflammation assessment, clinical populations