Abstract
Stress levels are surging, alongside the incidence of stress-related psychiatric disorders. Perhaps a related phenomenon, especially in urban areas, the human gut contains fewer bacterial species than ever before. Although the functional implications of this absence is unclear, one consequence may be reduced stress resilience. Preclinical and clinical evidence has shown how stress exposure can alter the gut microbiota and their metabolites, affecting host physiology. Also, stress-related shifts in the gut microbiota jeopardize tight junctions of the gut barrier. In this context, bacteria and bacterial products can translocate from the gut to the bloodstream, lymph nodes, and other organs, thereby modifying systemic inflammatory responses. Heightened circulating inflammation can be an etiological factor in stress-related psychiatric disorders, including some cases of depression. In this review, we detail preclinical and clinical evidence that traces these brain-to-gut-to-brain pathways that underlie stress-related psychiatric disorders and potentially affect their responsivity to conventional psychiatric medications. We also review evidence for interventions that modulate the gut microbiota (e.g., antibiotics, probiotics, prebiotics) to reduce stress responses and psychiatric symptoms. Lastly, we discuss challenges to translation and opportunities for innovations that could impact future psychiatric clinical practice.
Keywords: stress, Gut microbiota, Inflammation, intestinal permeability, depression
Introduction: Stress and Stress-Related Psychiatric Disorders
Worldwide, perceived stress continues to tick higher and higher, setting new records (1). Stress is inevitable, and therefore resilience is critical. Chronic and repetitive stressors increase the likelihood of psychiatric disorder onset in susceptible individuals, and yet many people successfully adapt to ongoing stressors (2). There are many reasons why people are unable to adapt, and for those who cannot adapt, psychiatric disorders can generate more stressors (e.g., conflict with people who do not understand the disorder) and can prolong the stressor’s psychological and physiological impact via anticipatory worry or post-event rumination (3–5) – core cognitive features of anxiety and depression. Although prior research conceptualized and measured stress differently, chronic or repetitive stressors that accumulate and uniquely interact with the individual’s context or history are most psychologically and physiologically detrimental (6). The stress response system, including the sympathetic-adrenal medullary (SAM) and hypothalamic-pituitary adrenal (HPA) axes, facilitates a functional response to time-limited threats that require a physical response (i.e., the classic fight-or-flight response), but the same response is potentially detrimental when the stressor is long-lasting and does not require a physical response. Stress-responsive systems are dysregulated in psychiatric disorders and may be possible etiological factors (7).
Both acute and chronic stress impact the immune system, which, over time, can result in psychological and behavioral changes. In humans, a 20-minute speech stressor increased inflammatory and antiviral gene expression via beta-adrenergic signaling (8). In contrast, chronic stress tracks with upregulated proinflammatory and downregulated anti-viral gene expression (9). Acute inflammatory rises can provoke a plethora of sickness behaviors – such as anhedonia, lack of appetite, and fatigue – paralleling depression symptoms (10). Indeed, higher basal inflammation may set the stage for depression, and vice versa (11). The transition from acute to chronic stress is not entirely clear (12), but acute stress that is repetitive may not allow for full recovery.
Social threats, especially those involving conflict, may be some of the most potent stressors (13). In humans, a speech for a panel of judges is a leading stress paradigm due to reproducible, steep endocrine and cytokine responses (14). We recently reported that those who had heightened inflammatory responses to this social-evaluative stressor had the greatest depressive symptom increases over time, especially if they reported frequent social stress in their daily lives (15). Although exaggerated inflammatory responses likely promoted survival during stressful situations throughout evolution, they are perhaps unnecessary and depressogenic in modern populations (16). Indeed, at least 25% of depression cases feature clinically elevated inflammation, evidenced by C-reactive protein (CRP, an acute phase protein produced during inflammation) of >3mg/L (17), with evidence of an anhedonia-prominent immune-mediated depression subtype (18). Moreover, circulating inflammatory markers have moderate to strong relationships with central nervous system (CNS) inflammatory markers in depressed patients (19). Although we primarily focus on depression in this review, stress and inflammation are also risk factors for the onset of other psychiatric disorders (e.g., schizophrenia) (20).
Failure to account for immune dysfunction as an etiological factor in some cases of depression may partially explain the high rates of treatment-resistant depression (21). Recognizing this etiological factor has led to novel therapeutic strategies, such as tumor necrosis factor-α antagonists for treatment-resistant depression, which are effective among those with elevated baseline inflammation (22). However, a newer frontier of immunopsychiatry involves the manipulation of the gut microbiota – comprised of fungi, archaea, viruses, yeast, and bacteria, the latter of which has received the most focus. The gut microbiota shape, and are shaped by immune function and psychological states (23). Emerging evidence implicates inflammation – locally in the gastrointestinal mucosa as well as systemically – and reduced integrity of the gut’s epithelial and mucosal lining (i.e., intestinal permeability, gut leakiness) as primary culprits connecting stress-induced gut microbiota shifts to psychiatric symptoms. In the Supplemental Material, we outline foundational stress-responsive pathways linking the gut, brain, and immune systems; the vagus nerve is particularly notable for its importance in both upstream and downstream signaling. Here we review key translational findings, first among animals and then humans, that (1) psychological stress impacts the gut microbiota, gut barrier integrity, metabolites, and systemic inflammation; and (2) these stress-related consequences contribute to poorer mental health.
Preclinical Findings: Downstream Effects from the Brain to the Gut
Over fifty years ago, stress’s effect on the gut microbiota emerged with the observation that rehoused mice had a lower abundance of beneficial Lactobacillus (24). Later, ex vivo and in vivo experiments demonstrated that stress hormones, particularly noradrenaline, could stimulate the growth of pathogenic bacteria (e.g., Escherichia coli) (25,26) and enhance adherence to mucosal tissue (27). One of the first systematic, culture-based in vivo studies investigated why infant rhesus monkeys often developed diarrhea after being weaned from their mothers (28). Infants showed significant declines in Lactobacillus three days after maternal separation. However, Lactobacillus abundance returned to baseline at the end of the week—an early indication of the microbiota’s resilience observed in other animal and human studies(29,30). Although this drop was not correlated with cortisol secretion, it was related to a more stressed behavioral phenotype, which itself predicted a higher risk of opportunistic infections (28). Lactobacillus reduction during stress is often reported in the literature, and it is consequential for immune function (31,32).
Subsequent research showed that stress could even affect the gut microbiota across generations. For example, in a culture-based study, an acoustic startle paradigm (10 minutes per day with three bursts of noise, five days per week for six weeks) led to lower abundances of Lactobacillus and Bifidobacterium in infants of prenatally-stressed rhesus monkey mothers versus non-stressed mothers for six months after birth (33). Again, there was a greater incidence of opportunistic microbial infection among infants of prenatally stressed mothers (40%) compared to those of non-stressed mothers (0%). These intergenerational stress effects on the gut microbiota, which have been replicated using more advanced 16S rRNA gene sequencing, may influence offspring colonic innervation and motility, HPA-axis stress responses, social behavior in males, and anxiety-like behavior in females (34–36). Intriguingly, germ-free mice that were separated from their mothers earlier in development, showed anxiety- and depressive-like behavior when their gut microbiome was reconstituted in adulthood. Importantly, anxiety- or depressive-like behavior did not develop after colonizing adult germ-free mice that were not separated from their mothers during development (37); thus, maternal separation may induce an intestinal environment that is more conducive to the growth or activity of bacteria associated with depression and anxiety. We discuss whether gut microbiota composition can be a biomarker of stress in the Supplemental Material.
Stress not only affects bacterial communities in the interior lumen of the intestine, but also in communities that are more closely associated with mucosal tissue. In mice exposed to a single two-hour social disruption (i.e., a dominant male intruder into a pre-established social cohort), diversity and relative abundances of mucosa-associated bacteria in the genera Parabacteroides and Lactobacillus, including L. reuteri, declined (38). These differences in mucosa-associated bacteria appear to be more consistent than differences in luminal bacteria, since exposure to a prolonged restraint stressor also affected mucosa-associated, but not luminal, diversity and Lactobacillus abundance (39).
These stress-induced alterations in the microbiome were accompanied by upregulated intestinal epithelial cell antimicrobial, proinflammatory, pro-oxidative gene expression (40). This is a common host response to bacterial contact with the intestinal epithelium. Indeed, in situ hybridization and a lectin-based mucus stain, showed that social defeat stress reduces the mucus barrier allowing bacteria to come into close contact with the epithelium (40,41). Reductions in intestinal mucus, and bacterial contact with the epithelium can lead to the translocation of intestinal bacteria (and bacterial products) to the interior of the body where they can exacerbate intestinal inflammation and/or trigger inflammatory responses throughout the body. The ensuing inflammatory response, which is often a low-grade inflammatory response, disrupts host physiology, including brain physiology, and is likely a key mechanism by which the gut microbiota can impact the brain and behavior responses to stress (41).
Translation to Humans: Downstream Effects from the Brain to the Gut
One of the first indications of brain-gut interactions in humans came from the clinical observation that stress seemed to trigger inflammatory bowel disease (IBD) flare-ups (42). Prospective evidence later showed that perceived stress, negative affect, and major life events, and not NSAIDs, antibiotics, or infections, predicted IBD flare-ups (43). Stress plays a major role in functional gastrointestinal disorders as well (44). Intestinal permeability1, low-grade systemic inflammation, and most notably mucosal inflammation characterize IBD, and intestinal epithelial cell shedding may foreshadow relapse (46). In one study, UC and Crohn’s disease patients had disproportionate abundance (up to 100-fold higher than healthy controls) of certain mucolytic bacteria (47). Psychological states are connected with these gastrointestinal manifestations: Compared to IBD patients without anxiety, depression, or high perceived stress, distressed patients have lower gut microbiota richness and diversity, as well as higher levels of potentially pathogenic bacteria (e.g., the genera Enterococcus, Streptococcus) and lower levels of beneficial microbes (e.g., Bifidobacterium) (48–50). Interestingly, transferring the gut microbiota of IBD patients with depression to specific pathogen free mice caused more severe colitis, endotoxin in the bloodstream, and depressive-like behavior, compared to those who received the microbiota of IBD patients without depression (49), showing that the gut microbiota play a central role in comorbid IBD and depression. In a multi-omics study of patients with ulcerative colitis, not only were distressed patients’ gut microbiota less diverse and rich, but microbiome differences were related to bile acid profiles and immune function (48). This correspondence between the gut microbiota and metabolome evident among mice (51) was also evident in another human IBD sample, which also showed differences in the metabolome related to bile acid secretion (52). Bile acids play a large role in shaping the gut microbiome (53), thus stress-associated alterations in bile acid profiles may have important effects on microbial community composition.
Even among healthy people, stress is associated with changes in the gut environment. To date, most human studies show non-experimental brain-to-gut associations – at best longitudinally. One line of research involved military-related stressors that are mostly physical in nature (e.g., four weeks of combat training) (54–56). These studies generally show increases in inflammation, gastrointestinal symptoms, stress, anxiety, depression, intestinal permeability, and even blood-brain-barrier (BBB) permeability in response to short-term military training (54–56). In one study, intestinal permeability increased in tandem with markers of inflammation, changes in gut microbiota composition, and metabolism following an extreme four-day hike through the Arctic (54). It is difficult to tease apart the unique roles of psychological versus physical stress in these studies because soldiers report heightened stress, anxiety, and depressive symptoms during these physical stressors; yet, those who report more gastrointestinal symptoms report the most distress during the physical challenge (55).
Outside the military paradigm, undergraduates’ levels of certain lactic acid-producing bacteria, such as Lactobacillus, decreased as academic stress mounted toward the end of the semester (57). Also, maternal prenatal stress tracks with offspring gut microbiota composition (58), similarly to animal studies (28). Although we are unaware of head-to-head comparisons between different stressor types and their effects on the gut-brain axis, early life stress and trauma, especially in relation to attachment figures, may canalize unhealthier trajectories because the microbiome is intimately involved in brain development (59). Interestingly, stressor-induced changes in the microbiome may be offset by parasympathetic activity. A recent report found that parasympathetic activity, as indexed by heart rate variability – indicative of vagal tone – was positively associated with human gut microbiota diversity and composition in women (60). Health behaviors and social relationships also affect the gut-brain axis (Supplemental Material).
Intestinal permeability, and associated translocation of bacteria or bacterial products, has emerged as a common finding in stress studies that assess the brain-gut axis. In addition to observational studies reviewed in the Supplemental Material, a handful of human studies have manipulated stress and measured intestinal permeability. For example, after immersing their hands in cold water, healthy women with moderate life stress, but not low stress, were more distressed, had decreased epithelial secretory ability, and greater intestinal permeability; however, there were no differences in the autonomic or hormonal response (61). A follow-up study showed that cold stress-induced intestinal permeability may be specific to women (62). In a more ecologically valid paradigm, healthy undergraduates who had significant salivary cortisol increases when they presented a bachelors or master’s thesis to an examination committee showed significant increases in intestinal permeability in a mast cell-dependent manner (63). This effect could be mimicked by administering corticotropin-releasing hormone (63), translating rodent findings of the centrality of mast cells and stress hormones in stress-induced gut leakiness (64–67).
Preclinical Evidence: Upstream Effects from the Gut to the Brain
Early findings that stress can alter gut bacteria led to studies examining whether these effects are bi-directional (i.e., whether gut bacteria could affect the stress response). Observations in germ-free mice helped to identify the importance of gut microbes for stress and anxiety. Compared to their conventional counterparts, germ-free mice have lower levels of anxiety-like behavior in basal conditions and following acute stress, even though they have exaggerated HPA-axis stress responses (68–71). Reconstitution with Bifidobacterium infantis can normalize, whereas Escherichia coli can enhance, plasma adrenocorticotropic hormone and corticosterone responses (68). However, reconstitution is not able to restore all aspects of CNS neurotransmission. For example, hippocampal serotonin remained low after recolonization even though serotonin is influenced by the gut microbiota and serotonin’s precursor tryptophan returned to baseline in the periphery of reconstituted mice (69). Other research also shows a tryptophan-serotonin imbalance in germ-free mice (51), illustrating the important role of gut bacteria in tryptophan metabolism and serotonin homeostasis. In addition, gut bacteria shape the adrenal and pituitary gland’s stress response, attenuating genetic expression of a glucocorticoid receptor sensitivity modulator in the pituitary and of steroidogenesis and catecholamine synthesis in the adrenal gland (72).
Germ-free mice have also been useful for illustrating the impact that stress-induced changes to the microbiome can have on host physiology. For example, compared to dominant mice, submissive mice have significantly lower gut microbiota diversity, a different bacterial composition, lower body weight, less white adipose tissue, and greater inflammation (73). When the submissive microbiome was transplanted into germ-free mice, recipients developed the submissive behavioral profile (i.e., anti-social, depressive-like behavior) and physiological phenotype (e.g., smaller adipocytes). One step further, on the cusp of translational research were the findings by Kelly et al. showing that transferring gut microbiota from depressed humans, which were less diverse than that of non-depressed humans, to germ-free mice initiated depressive-like behavior (74), confirming the behavioral impact of the microbiome.
The mechanisms by which the microbiome may impact behavior during stress are complex and multifactorial but can involve the immune system. In mice, repeated social defeat leads to anxiety-like behavior and cognitive deficits, which are dependent upon immune activation (75). Neuronal release of interleukin (IL)-1β leads to microglial activation and subsequent recruitment of inflammatory monocytes to the brain (75). We have found social defeat fails to increase inflammatory cytokines, as well as hippocampal neuronal and microglial activation, when mice are treated with an antibiotic cocktail to affect the microbiota (41). Social defeat led to mucus depletion and increased lipopolysaccharide (LPS) binding protein (LBP) (41), which can occur when intestinal bacteria (or bacterial components like LPS) translocate from the intestinal lumen to the interior of the body through a leaky gut. LBP binding to CD14, a receptor on the surface of leukocytes which leads to increased inflammatory cytokines, was necessary for social defeat-induced increases in hippocampal neuronal and microglial activation (41). Although this study focused on the dorsal hippocampus, others have found that the microbiota from stressed mice drive increases in Il-1β in the ventral hippocampus and increased depressive-like behavior (76). These studies illustrate that during stress, the microbiota contribute to hippocampal neuroinflammation related to anxiety and depressive-like behavior.
Findings that the microbiota contribute to stressor-induced behavioral changes have led to translational studies testing whether targeting the microbiome can improve behavioral responses. Pre-stress gut microbiota modulation via probiotics, prebiotics, and antibiotics can alter the behavioral response to stress, including stress resilience. In mice exposed to chronic social defeat stress, about one-third are resilient (i.e., do not develop depressive-like behavior). These resilient mice have higher abundances of Bifidobacterium compared to non-resilient mice. Interestingly, Bifidobacterium supplementation leads to increased resilience (77). Multiple studies have demonstrated that pre-stress antibiotic treatment can block stress-related depressive- and anxiety-like behavior (78,79). Similarly, prebiotic fiber can modify behavioral responses to stress. Among male mice, three weeks of fructooligosaccharide and galactooligosaccharide supplementation increased short-chain fatty acid (SCFA) concentration, boosted serotonin levels in the prefrontal cortex, changed gene expression in the hippocampus and hypothalamus, and reduced chronic social stress-related increases in glucocorticoids and depressive- and anxiety-like behavior (80). Human milk oligosaccharides have also been tested in mice, and after two weeks of supplementation, the colonic mucosa-associated microbiota community structure was resilient to social disruption stress. Consistent with this observation, the supplemented mice had normalized anxiety-like behavior and even improved neuronal maturation in the dentate gyrus (81) – a brain structure implicated in depression and anxiety. See (82) for a comprehensive review of probiotic and prebiotic supplementation.
Translation to Humans: Upstream Effects from the Gut to Brain
It is now well recognized that gut microbiota and metabolites are associated with human mood and behavioral differences. For example, in a gut microbiome-wide association study of depressive symptoms, developed with the Rotterdam Study cohort (N=1,054) and validated in the Amsterdam HELIUS cohort (N=1,539), not only was microbiota diversity lower, but depressed individuals had significantly different abundances of bacteria that synthesize glutamate, butyrate, serotonin, and GABA, compared to healthy controls (83). Indeed, evidence has accumulated that key features of the metabolome, including pathways involved in neurotransmission, glutamatergic and energy metabolism, reliably distinguish patients with unipolar and bipolar depression from each other and from healthy controls (84). These depression-related metabolome signatures interact with the immune system. For example, glutamate via NMDA receptors can increase neuronal proinflammatory cytokine expression (85), and SCFAs like butyrate dampen inflammatory responses (86).
Perhaps not surprisingly, findings that the microbiome is associated with mood have led to studies testing whether probiotics and prebiotics can improve mood. Multiple meta-analyses suggest that probiotic administration, especially multispecies supplementation, may modestly reduce depressive symptoms among those with mild to moderate depression, but not among healthy people (87,88). However, despite supportive animal studies, to date, the evidence is not strong for probiotic anxiolytic effects in humans (89–91). The reasons for the discrepant findings are not exactly clear, but the laboratory animal microbiome is significantly different than the human microbiome (92). Thus, microbiome contributions to probiotic beneficial effects likely differ in laboratory animals and humans. Moreover, probiotic dosing in laboratory animals is often similar to dosing used in humans, even though laboratory animals are much smaller (93). Fewer studies have focused on prebiotics, but when pooling effects from the small number of extant human studies – most of them not psychiatric samples – there is not overwhelming support for the idea that prebiotics can improve clinical depression or anxiety (94).
Probiotics, prebiotics, or synbiotics (the combination of probiotics and prebiotics) may have more beneficial effects when administered as adjunctive treatments to conventional medication (e.g., SSRIs). The small number of trials to date have shown that these interventions are more effective than first-line treatment alone or with placebo in generalized anxiety disorder and major depressive disorder (95). In contrast, adjunctive probiotic treatment increased tolerability of first-line treatments in schizophrenia but had no effect on clinical outcomes (95). Interestingly, a recent meta-analysis of three randomized, placebo-controlled trials in humans showed that the tetracycline antibiotic minocycline reduced depressive symptoms (effect size = −0.78) (96); however, it is not yet clear whether shifts in the gut microbiota mediate this antidepressant effect, as minocycline easily crosses the BBB and reduces the proinflammatory potential of microglia (97).
There is accumulating evidence that gut leakiness is evident in stress-related psychiatric disorders. Several cross-sectional studies have found evidence for greater intestinal permeability among depressed patients, compared to healthy controls (98–101). In fact, intestinal permeability may be especially elevated in persistent and severe depression (100,102). Further, metagenome sequencing showed that anxious and depressed patients have increased abundance of bacterial genes related to mucus degradation, as well as endotoxin biosynthesis, compared to their peers without a psychiatric disorder (98). Gut leakiness, however, may not be unique to depression: Meta-analytic evidence indicates that a wide range of leaky gut markers are higher among those with severe mental illness and chronic fatigue, compared to healthy controls, and that these markers positively track with sickness behaviors (103). Thus, intestinal permeability may be a transdiagnostic risk factor for sickness behaviors. In terms of prospective risk, we found that LBP predicted depressive symptoms one and two years later among 315 women, but the reverse relationship was non-significant. Leaky gut’s relationship with future depressive symptoms was particularly strong among those with high levels of circulating inflammation at baseline (104), indicating that gut leakiness could be a unique risk factor for development of inflammation-driven depressive symptoms.
There are a few notable studies for administering probiotics to healthy humans, manipulating stress exposure, and measuring resultant gut leakiness as well as self-reported and physiological stress. In one small, randomized double-blind, placebo-controlled trial, healthy young males and females who received twice-daily Lactobacillus rhamnosus milk for one month prior to a thesis defense showed lower intestinal permeability, anxiety, and perceived stress in comparison to individuals that received acidified milk as a control. Interestingly, there were no differences in cortisol, suggesting the probiotics may affect permeability independently from effects on the endocrine response to stress (105). These results contrast with another RCT among healthy young males, which showed that eight weeks of L. rhamnosus did not impact mood, anxiety, stress, sleep quality, cognitive performance, inflammation, or salivary cortisol responses to a socially-evaluated cold pressor test (106). This lack of effects in males suggest that future work should examine sex as a potential moderator. Additionally, probiotic strain specificity is critical, as there is considerable variability.
Conclusion
Preclinical and clinical evidence demonstrate that chronic and repetitive stressors – particularly social stressors — reduce the diversity and the relative abundance of beneficial bacteria in the intestine. These stressors also lead to altered metabolomic profiles that are associated with dysregulation of whole-body energy dynamics, greater intestinal permeability and translocation of bacteria and bacterial products across the gut barrier, and enhanced mucosal and circulating inflammation. The heightened circulating inflammation is particularly relevant for stress-related psychiatric disorders, which often coincide with enhanced inflammatory responses. Thus, targeting the gut microbiota is a promising approach to promote physiological stress resilience that may in turn foster psychological resilience and reduce the incidence of stress-related psychiatric disorders.
That said, experimental evidence in humans is nascent, and there are several challenges to translating preclinical findings. It is currently not clear what constitutes a healthy human gut microbiome, as it is highly variable between individuals (107,108), and many microbial genes and functions are not yet characterized (109). In addition, the type and “dose” of stress exposure that affects the gut microbiota remains unclear, as preclinical stress paradigms (e.g., social defeat) do not precisely parallel clinical stress paradigms. Moreover, during stressful periods, human dietary patterns change and it is difficult to account for dietary variation in humans when stressed (unlike preclinical studies utilizing standardized chow). Finally, there are substantial cross-cultural differences in human physiological responses to stress (110), which deserve further consideration in the gut-brain axis literature as most studies have focused on Western populations. The Supplemental Material outlines future directions.
Overall, the gut microbiota is a critical element in stress-related psychiatric disorders, as microbial genes in the human body outnumber human genes by 150-fold(111). The prevalence of psychiatric disorders that do not respond to conventional treatments calls for a more nuanced and in-depth understanding into how peripheral physiology, including non-human cells in the gut, may drive certain psychiatric symptoms and treatment responses in the context of stress.
Supplementary Material
Figure 1.
Stress-related changes in the gut affect the immune system, peripheral organs, and brain. (A) Psychological stress has downstream consequences for the gut (e.g., communicated via the vagus nerve [depicted in yellow] hypothalamic-pituitary adrenal [HPA] axis, and sympathetic nervous system). Notably, environmental stress, such as lower environmental biodiversity and poor dietary habits, can directly impact the gut. (B) During stress, the relative abundance of beneficial bacteria in the lumen often decrease, as does overall richness and diversity. Mucosal mast cell degranulation can damage the epithelial cell lining and compromise barrier integrity. (C) Gut bacteria and bacterial products (like lipopolysaccharide) can then translocate into the bloodstream, spleen, and lymph nodes (lymphatic system depicted in green), priming monocytes and triggering a systemic inflammatory response. (D) Higher levels of peripheral inflammation can also affect blood brain barrier permeability and increase neuroinflammation. These stress-related physiological changes can, in turn, enhance the harmful effects of subsequent stressors, decrease resilience, and heighten risk for psychiatric disorders.
Acknowledgements
This work was supported in part by 4R33MH108167-03.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflicts of Interest
MTB is a Scientific Cofounder and owns stock options in Scioto Biosciences. AAM reports no biomedical financial interests or potential conflicts of interest.
1 Note that there are several ways to measure and quantify intestinal permeability in humans, and the pros and cons of these measurements is beyond the scope of this article. See (45) for a discussion of some techniques in humans.
References
- 1.Gallup Global Emotions Report (2022): The Gallup Organization. [Google Scholar]
- 2.Kalisch R, Baker DG, Basten U, Boks MP, Bonanno GA, Brummelman E, et al. (2017): The resilience framework as a strategy to combat stress-related disorders. Nature human behaviour 1: 784–790. [DOI] [PubMed] [Google Scholar]
- 3.Brosschot JF, Pieper S, Thayer JF (2005): Expanding stress theory: Prolonged activation and perseverative cognition. Psychoneuroendocrinology 30: 1043–1049. [DOI] [PubMed] [Google Scholar]
- 4.Conway CC, Hammen C, Brennan PA (2012): Expanding stress generation theory: test of a transdiagnostic model. Journal of Abnormal Psychology 121: 754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ottaviani C, Thayer JF, Verkuil B, Lonigro A, Medea B, Couyoumdjian A, Brosschot JF (2016): Physiological concomitants of perseverative cognition: A systematic review and meta-analysis. Psychological bulletin 142: 231. [DOI] [PubMed] [Google Scholar]
- 6.Epel ES, Crosswell AD, Mayer SE, Prather AA, Slavich GM, Puterman E, Mendes WB (2018): More than a feeling: A unified view of stress measurement for population science. Frontiers in neuroendocrinology 49: 146–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Turner AI, Smyth N, Hall SJ, Torres SJ, Hussein M, Jayasinghe SU, et al. (2020): Psychological stress reactivity and future health and disease outcomes: A systematic review of prospective evidence. Psychoneuroendocrinology 104599. [DOI] [PubMed] [Google Scholar]
- 8.MacCormack JK, Gaudier-Diaz MM, Armstrong-Carter EL, Arevalo JM, Meltzer-Brody S, Sloan EK, et al. (2021): Beta-adrenergic blockade blunts inflammatory and antiviral/antibody gene expression responses to acute psychosocial stress. Neuropsychopharmacology 46: 756–762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Cole SW (2019): The conserved transcriptional response to adversity. Current opinion in behavioral sciences 28: 31–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lasselin J (2021): Back to the future of psychoneuroimmunology: Studying inflammation-induced sickness behavior. Brain, Behavior, & Immunity-Health 18: 100379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mac Giollabhui N, Ng TH, Ellman LM, Alloy LB (2020): The longitudinal associations of inflammatory biomarkers and depression revisited: systematic review, meta-analysis, and meta-regression. Molecular Psychiatry 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Rohleder N (2019): Stress and inflammation–The need to address the gap in the transition between acute and chronic stress effects. Psychoneuroendocrinology 105: 164–171. [DOI] [PubMed] [Google Scholar]
- 13.Slavich GM (2020): Social safety theory: a biologically based evolutionary perspective on life stress, health, and behavior. Annual review of clinical psychology 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kirschbaum C, Pirke K-M, Hellhammer DH (1993): The ‘Trier Social Stress Test’–a tool for investigating psychobiological stress responses in a laboratory setting [no. 1–2]. Neuropsychobiology 28: 76–81. [DOI] [PubMed] [Google Scholar]
- 15.Madison AA, Andridge R, Shrout MR, Renna ME, Bennett JM, Jaremka LM, et al. (2021): Frequent interpersonal stress and inflammatory reactivity predict depressive symptom increases: Two tests of the social signal transduction theory of depression. Psychological Science 33: 152–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Raison CL, Miller A (2013): The evolutionary significance of depression in Pathogen Host Defense (PATHOS-D) [no. 1]. Molecular psychiatry 18: 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Osimo EF, Pillinger T, Rodriguez IM, Khandaker GM, Pariante CM, Howes OD (2020): Inflammatory markers in depression: a meta-analysis of mean differences and variability in 5,166 patients and 5,083 controls. Brain, behavior, and immunity 87: 901–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Haroon E, Chen X, Li Z, Patel T, Woolwine BJ, Hu XP, et al. (2018): Increased inflammation and brain glutamate define a subtype of depression with decreased regional homogeneity, impaired network integrity, and anhedonia. Translational psychiatry 8: 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Felger JC, Haroon E, Patel TA, Goldsmith DR, Wommack EC, Woolwine BJ, et al. (2018): What does plasma CRP tell us about peripheral and central inflammation in depression? Molecular psychiatry 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Howes O, McCutcheon R (2017): Inflammation and the neural diathesis-stress hypothesis of schizophrenia: a reconceptualization. Translational psychiatry 7: e1024–e1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Chamberlain SR, Cavanagh J, De Boer P, Mondelli V, Jones DN, Drevets WC, et al. (2019): Treatment-resistant depression and peripheral C-reactive protein. The British Journal of Psychiatry 214: 11–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Raison CL, Rutherford RE, Woolwine BJ, Shuo C, Schettler P, Drake DF, et al. (2013): A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers [no. 1]. JAMA psychiatry 70: 31–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Powell N, Walker MM, Talley NJ (2017): The mucosal immune system: master regulator of bidirectional gut–brain communications. Nature reviews Gastroenterology & hepatology 14: 143–159. [DOI] [PubMed] [Google Scholar]
- 24.Schaedler RW, Dubos RJ (1962): The fecal flora of various strains of mice. Its bearing on their susceptibility to endotoxin. The Journal of Experimental Medicine 115: 1149–1160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Lyte M, Frank CD, Green BT (1996): Production of an autoinducer of growth by norepinephrine cultured Escherichia coli O157: H7. FEMS microbiology letters 139: 155–159. [DOI] [PubMed] [Google Scholar]
- 26.Lyte M, Bailey MT (1997): Neuroendocrine–bacterial interactions in a neurotoxin-induced model of trauma. Journal of Surgical Research 70: 195–201. [DOI] [PubMed] [Google Scholar]
- 27.Vlisidou I, Lyte M, Van Diemen PM, Hawes P, Monaghan P, Wallis TS, Stevens MP (2004): The neuroendocrine stress hormone norepinephrine augments Escherichia coli O157: H7-induced enteritis and adherence in a bovine ligated ileal loop model of infection. Infection and immunity 72: 5446–5451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bailey MT, Coe CL (1999): Maternal separation disrupts the integrity of the intestinal microflora in infant rhesus monkeys. Developmental Psychobiology: The Journal of the International Society for Developmental Psychobiology 35: 146–155. [PubMed] [Google Scholar]
- 29.Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R (2012): Diversity, stability and resilience of the human gut microbiota. Nature 489: 220–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Bailey MT, Dowd SE, Galley JD, Hufnagle AR, Allen RG, Lyte M (2011): Exposure to a social stressor alters the structure of the intestinal microbiota: implications for stressor-induced immunomodulation [no. 3]. Brain, behavior, and immunity 25: 397–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Galley JD, Bailey MT (2014): Impact of stressor exposure on the interplay between commensal microbiota and host inflammation. Gut microbes 5: 390–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mackos AR, Galley JD, Eubank TD, Easterling RS, Parry NM, Fox JG, et al. (2016): Social stress-enhanced severity of Citrobacter rodentium-induced colitis is CCL2-dependent and attenuated by probiotic Lactobacillus reuteri. Mucosal immunology 9: 515–526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Bailey MT, Lubach GR, Coe CL (2004): Prenatal stress alters bacterial colonization of the gut in infant monkeys. Journal of pediatric gastroenterology and nutrition 38: 414–421. [DOI] [PubMed] [Google Scholar]
- 34.Golubeva AV, Crampton S, Desbonnet L, Edge D, O’Sullivan O, Lomasney KW, et al. (2015): Prenatal stress-induced alterations in major physiological systems correlate with gut microbiota composition in adulthood. Psychoneuroendocrinology 60: 58–74. [DOI] [PubMed] [Google Scholar]
- 35.Gur TL, Shay L, Palkar AV, Fisher S, Varaljay VA, Dowd S, Bailey MT (2017): Prenatal stress affects placental cytokines and neurotrophins, commensal microbes, and anxiety-like behavior in adult female offspring. Brain, behavior, and immunity 64: 50–58. [DOI] [PubMed] [Google Scholar]
- 36.Gur TL, Palkar AV, Rajasekera T, Allen J, Niraula A, Godbout J, Bailey MT (2019): Prenatal stress disrupts social behavior, cortical neurobiology and commensal microbes in adult male offspring. Behavioural brain research 359: 886–894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.De Palma G, Blennerhassett P, Lu J, Deng Y, Park A, Green W, et al. (2015): Microbiota and host determinants of behavioural phenotype in maternally separated mice. Nature communications 6: 7735. [DOI] [PubMed] [Google Scholar]
- 38.Galley JD, Nelson MC, Yu Z, Dowd SE, Walter J, Kumar PS, et al. (2014): Exposure to a social stressor disrupts the community structure of the colonic mucosa-associated microbiota [no. 1]. BMC microbiology 14: 189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Galley JD, Yu Z, Kumar P, Dowd SE, Lyte M, Bailey MT (2014): The structures of the colonic mucosa-associated and luminal microbial communities are distinct and differentially affected by a prolonged murine stressor [no. 6]. Gut microbes 5: 748–760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Allen JM, Mackos AR, Jaggers RM, Brewster PC, Webb M, Lin C-H, et al. (2022): Psychological stress disrupts intestinal epithelial cell function and mucosal integrity through microbe and host-directed processes. Gut Microbes 14: 2035661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Jaggers RM, DiSabato DJ, Loman BR, Kontic D, Spencer KD, Allen JM, et al. (2022): Stressor-induced reduction in cognitive behavior is associated with impaired colonic mucus layer integrity and is dependent upon the LPS-binding protein receptor CD14. Journal of Inflammation Research 1617–1635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Mckegney PF, Gordon RO, Levine SM (1970): A psychosomatic comparison of patients with ulcerative colitis and Crohn’s disease. Psychosomatic Medicine 32: 153–166. [DOI] [PubMed] [Google Scholar]
- 43.Bernstein CN, Singh S, Graff LA, Walker JR, Miller N, Cheang M (2010): A prospective population-based study of triggers of symptomatic flares in IBD. Official journal of the American College of Gastroenterology| ACG 105: 1994–2002. [DOI] [PubMed] [Google Scholar]
- 44.Qin H-Y, Cheng C-W, Tang X-D, Bian Z-X (2014): Impact of psychological stress on irritable bowel syndrome. World journal of gastroenterology: WJG 20: 14126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Camilleri M (2019): Leaky gut: mechanisms, measurement and clinical implications in humans. Gut 68: 1516–1526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kiesslich 1R, Duckworth C, Moussata D, Gloeckner A, Lim L, Goetz M, et al. (2012): Local barrier dysfunction identified by confocal laser endomicroscopy predicts relapse in inflammatory bowel disease. Gut 61: 1146–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Png CW, Lindén SK, Gilshenan KS, Zoetendal EG, McSweeney CS, Sly LI, et al. (2010): Mucolytic bacteria with increased prevalence in IBD mucosa augmentin vitroutilization of mucin by other bacteria. Official journal of the American College of Gastroenterology| ACG 105: 2420–2428. [DOI] [PubMed] [Google Scholar]
- 48.Yuan X, Chen B, Duan Z, Xia Z, Ding Y, Chen T, et al. (2021): Depression and anxiety in patients with active ulcerative colitis: crosstalk of gut microbiota, metabolomics and proteomics. Gut Microbes 13: 1987779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jang H-M, Kim J-K, Joo M-K, Shin Y-J, Lee CK, Kim H-J, Kim D-H (2021): Transplantation of fecal microbiota from patients with inflammatory bowel disease and depression alters immune response and behavior in recipient mice. Scientific Reports 11: 20406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Humbel F, Rieder JH, Franc Y, Juillerat P, Scharl M, Misselwitz B, et al. (2020): Association of alterations in intestinal microbiota with impaired psychological function in patients with inflammatory bowel diseases in remission. Clinical gastroenterology and hepatology 18: 2019–2029. [DOI] [PubMed] [Google Scholar]
- 51.Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, Siuzdak G (2009): Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites [no. 10]. Proceedings of the national academy of sciences 106: 3698–3703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Vila AV, Hu S, Andreu-Sánchez S, Collij V, Jansen BH, Augustijn HE, et al. (2023): Faecal metabolome and its determinants in inflammatory bowel disease. Gut. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Ridlon JM, Kang DJ, Hylemon PB, Bajaj JS (2014): Bile acids and the gut microbiome. Current opinion in gastroenterology 30: 332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Karl JP, Margolis LM, Madslien EH, Murphy NE, Castellani JW, Gundersen Y, et al. (2017): Changes in intestinal microbiota composition and metabolism coincide with increased intestinal permeability in young adults under prolonged physiological stress [no. 6]. American Journal of Physiology-Gastrointestinal and Liver Physiology 312: G559–G571. [DOI] [PubMed] [Google Scholar]
- 55.Li X, Kan E, Lu J, Cao Y, Wong R, Keshavarzian A, Wilder‐ Smith C (2013): Combattraining increases intestinal permeability, immune activation and gastrointestinal symptoms in soldiers [no. 8]. Alimentary pharmacology & therapeutics 37: 799–809. [DOI] [PubMed] [Google Scholar]
- 56.Varanoske AN, McClung HL, Sepowitz JJ, Halagarda CJ, Farina EK, Berryman CE, et al. (2022): Stress and the gut-brain axis: Cognitive performance, mood state, and biomarkers of blood-brain barrier and intestinal permeability following severe physical and psychological stress. Brain, Behavior, and Immunity 101: 383–393. [DOI] [PubMed] [Google Scholar]
- 57.Knowles SR, Nelson EA, Palombo EA (2008): Investigating the role of perceived stress on bacterial flora activity and salivary cortisol secretion: a possible mechanism underlying susceptibility to illness. Biological psychology 77: 132–137. [DOI] [PubMed] [Google Scholar]
- 58.Zijlmans MA, Korpela K, Riksen-Walraven JM, de Vos WM, de Weerth C (2015): Maternal prenatal stress is associated with the infant intestinal microbiota. Psychoneuroendocrinology 53: 233–245. [DOI] [PubMed] [Google Scholar]
- 59.Collins SM, Surette M, Bercik P (2012): The interplay between the intestinal microbiota and the brain. Nature Reviews Microbiology 10: 735–742. [DOI] [PubMed] [Google Scholar]
- 60.Mörkl S, Oberascher A, Tatschl JM, Lackner S, Bastiaanssen TF, Butler MI, et al. (2022): Cardiac vagal activity is associated with gut-microbiome patterns in women—An exploratory pilot study. Dialogues in Clinical Neuroscience 24: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Alonso C, Guilarte M, Vicario M, Ramos L, Ramadan Z, Antolín M, et al. (2008): Maladaptive intestinal epithelial responses to life stress may predispose healthy women to gut mucosal inflammation. Gastroenterology 135: 163–172. [DOI] [PubMed] [Google Scholar]
- 62.Alonso C, Guilarte M, Vicario M, Ramos L, Rezzi S, Martínez C, et al. (2012): Acute experimental stress evokes a differential gender‐ determined increase in human intestinal macromolecular permeability. Neurogastroenterology & Motility 24: 740–e349. [DOI] [PubMed] [Google Scholar]
- 63.Vanuytsel T, van Wanrooy S, Vanheel H, Vanormelingen C, Verschueren S, Houben E, et al. (2013): Psychological stress and corticotropin-releasing hormone increase intestinal permeability in humans by a mast cell-dependent mechanism. Gut gutjnl-2013. [DOI] [PubMed] [Google Scholar]
- 64.Meddings JB, Swain M (2000): Environmental stress–induced gastrointestinal permeability is mediated by endogenous glucocorticoids in the rat. Gastroenterology 119: 1019–1028. [DOI] [PubMed] [Google Scholar]
- 65.Kiliaan AJ, Saunders PR, Bijlsma PB, Berin MC, Taminiau JA, Groot JA, Perdue MH (1998): Stress stimulates transepithelial macromolecular uptake in rat jejunum. American Journal of Physiology-Gastrointestinal and Liver Physiology 275: G1037–G1044. [DOI] [PubMed] [Google Scholar]
- 66.Söderholm JD, Yang P, Ceponis P, Vohra A, Riddell R, Sherman PM, Perdue MH (2002): Chronic stress induces mast cell–dependent bacterial adherence and initiates mucosal inflammation in rat intestine. Gastroenterology 123: 1099–1108. [DOI] [PubMed] [Google Scholar]
- 67.Vicario M, Guilarte M, Alonso C, Yang P, Martínez C, Ramos L, et al. (2010): Chronological assessment of mast cell-mediated gut dysfunction and mucosal inflammation in a rat model of chronic psychosocial stress. Brain, behavior, and immunity 24: 1166–1175. [DOI] [PubMed] [Google Scholar]
- 68.Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu X, et al. (2004): Postnatal microbial colonization programs the hypothalamic–pituitary–adrenal system for stress response in mice. The Journal of physiology 558: 263–275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Clarke G, Grenham S, Scully P, Fitzgerald P, Moloney R, Shanahan F, et al. (2013): The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Molecular psychiatry 18: 666–673. [DOI] [PubMed] [Google Scholar]
- 70.Crumeyrolle-Arias M, Jaglin M, Bruneau A, Vancassel S, Cardona A, Daugé V, et al. (2014): Absence of the gut microbiota enhances anxiety-like behavior and neuroendocrine response to acute stress in rats. Psychoneuroendocrinology 42: 207–217. [DOI] [PubMed] [Google Scholar]
- 71.Neufeld K, Kang N, Bienenstock J, Foster JA (2011): Reduced anxiety‐ like behavior and central neurochemical change in germ‐ free mice. Neurogastroenterology & Motility 23: 255–e119. [DOI] [PubMed] [Google Scholar]
- 72.Vodička M, Ergang P, Hrnčíř T, Mikulecká A, Kvapilová P, Vagnerová K, et al. (2018): Microbiota affects the expression of genes involved in HPA axis regulation and local metabolism of glucocorticoids in chronic psychosocial stress. Brain, behavior, and immunity 73: 615–624. [DOI] [PubMed] [Google Scholar]
- 73.Agranyoni O, Meninger-Mordechay S, Uzan A, Ziv O, Salmon-Divon M, Rodin D, et al. (2021): Gut microbiota determines the social behavior of mice and induces metabolic and inflammatory changes in their adipose tissue. NPJ biofilms and microbiomes 7: 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Kelly JR, Borre Y, O’Brien C, Patterson E, El Aidy S, Deane J, et al. (2016): Transferring the blues: depression-associated gut microbiota induces neurobehavioural changes in the rat. Journal of psychiatric research 82: 109–118. [DOI] [PubMed] [Google Scholar]
- 75.McKim DB, Weber MD, Niraula A, Sawicki CM, Liu X, Jarrett BL, et al. (2018): Microglial recruitment of IL-1β-producing monocytes to brain endothelium causes stress-induced anxiety. Molecular psychiatry 23: 1421–1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Pearson-Leary J, Zhao C, Bittinger K, Eacret D, Luz S, Vigderman AS, et al. (2020): The gut microbiome regulates the increases in depressive-type behaviors and in inflammatory processes in the ventral hippocampus of stress vulnerable rats. Molecular psychiatry 25: 1068–1079. [DOI] [PubMed] [Google Scholar]
- 77.Yang C, Fujita Y, Ren Q, Ma M, Dong C, Hashimoto K (2017): Bifidobacterium in the gut microbiota confer resilience to chronic social defeat stress in mice. Scientific reports 7: 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wang S, Qu Y, Chang L, Pu Y, Zhang K, Hashimoto K (2020): Antibiotic-induced microbiome depletion is associated with resilience in mice after chronic social defeat stress. Journal of affective disorders 260: 448–457. [DOI] [PubMed] [Google Scholar]
- 79.Meng C, Feng S, Hao Z, Dong C, Liu H (2022): Antibiotics exposure attenuates chronic unpredictable mild stress-induced anxiety-like and depression-like behavior. Psychoneuroendocrinology 136: 105620. [DOI] [PubMed] [Google Scholar]
- 80.Burokas A, Arboleya S, Moloney RD, Peterson VL, Murphy K, Clarke G, et al. (2017): Targeting the microbiota-gut-brain axis: prebiotics have anxiolytic and antidepressantlike effects and reverse the impact of chronic stress in mice [no. 7]. Biological psychiatry 82: 472–487. [DOI] [PubMed] [Google Scholar]
- 81.Tarr AJ, Galley JD, Fisher SE, Chichlowski M, Berg BM, Bailey MT (2015): The prebiotics 3′ Sialyllactose and 6′ Sialyllactose diminish stressor-induced anxiety-like behavior and colonic microbiota alterations: Evidence for effects on the gut–brain axis. Brain, behavior, and immunity 50: 166–177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sarkar A, Lehto SM, Harty S, Dinan TG, Cryan JF, Burnet PW (2016): Psychobiotics and the manipulation of bacteria–gut–brain signals [no. 11]. Trends in neurosciences 39: 763–781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Radjabzadeh D, Bosch JA, Uitterlinden AG, Zwinderman AH, Ikram MA, van Meurs JB, et al. (2022): Gut microbiome-wide association study of depressive symptoms. Nature Communications 13: 7128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.MacDonald K, Krishnan A, Cervenka E, Hu G, Guadagno E, Trakadis Y (2019): Biomarkers for major depressive and bipolar disorders using metabolomics: A systematic review. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics 180: 122–137. [DOI] [PubMed] [Google Scholar]
- 85.Kitaoka Y, Munemasa Y, Nakazawa T, Ueno S (2007): NMDA-induced interleukin-1β expression is mediated by nuclear factor-kappa B p65 in the retina. Brain research 1142: 247–255. [DOI] [PubMed] [Google Scholar]
- 86.Segain J, De La Blétiere DR, Bourreille A, Leray V, Gervois N, Rosales C, et al. (2000): Butyrate inhibits inflammatory responses through NFκB inhibition: implications for Crohn’s disease. Gut 47: 397–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Ng QX, Peters C, Ho CYX, Lim DY, Yeo W-S (2018): A meta-analysis of the use of probiotics to alleviate depressive symptoms. Journal of affective disorders 228: 13–19. [DOI] [PubMed] [Google Scholar]
- 88.Goh KK, Liu Y-W, Kuo P-H, Chung Y-CE, Lu M-L, Chen C-H (2019): Effect of probiotics on depressive symptoms: A meta-analysis of human studies. Psychiatry research 282: 112568. [DOI] [PubMed] [Google Scholar]
- 89.Reis DJ, Ilardi SS, Punt SE (2018): The anxiolytic effect of probiotics: A systematic review and meta-analysis of the clinical and preclinical literature. PloS one 13: e0199041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Vitellio P, Chira A, De Angelis M, Dumitrascu DL, Portincasa P (2020): Probiotics in Psychosocial Stress and Anxiety. A Systematic Review. Journal of Gastrointestinal & Liver Diseases 29. [DOI] [PubMed] [Google Scholar]
- 91.Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, et al. (2011): Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proceedings of the National Academy of Sciences 201102999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Nguyen TLA, Vieira-Silva S, Liston A, Raes J (2015): How informative is the mouse for human gut microbiota research? Disease models & mechanisms 8: 1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Casteleyn C, Rekecki A, Van der Aa A, Simoens P, Van Den Broeck W (2010): Surface area assessment of the murine intestinal tract as a prerequisite for oral dose translation from mouse to man. Laboratory animals 44: 176–183. [DOI] [PubMed] [Google Scholar]
- 94.Liu RT, Walsh RF, Sheehan AE (2019): Prebiotics and probiotics for depression and anxiety: a systematic review and meta-analysis of controlled clinical trials. Neuroscience & Biobehavioral Reviews 102: 13–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Forth E, Buehner B, Storer A, Sgarbossa C, Milev R, Meyyappan AC (2023): Systematic review of probiotics as an adjuvant treatment for psychiatric disorders. Frontiers in Behavioral Neuroscience 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Rosenblat JD, McIntyre RS (2018): Efficacy and tolerability of minocycline for depression: a systematic review and meta-analysis of clinical trials. Journal of affective disorders 227: 219–225. [DOI] [PubMed] [Google Scholar]
- 97.Henry CJ, Huang Y, Wynne A, Hanke M, Himler J, Bailey MT, et al. (2008): Minocycline attenuates lipopolysaccharide (LPS)-induced neuroinflammation, sickness behavior, and anhedonia. Journal of neuroinflammation 5: 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Stevens BR, Goel R, Seungbum K, Richards EM, Holbert RC, Pepine CJ, Raizada MK (2018): Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression [no. 8]. Gut 67: 1555–1557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Mass M, Kubera M, Leunis J-C (2008): The gut-brain barrier in major depression: intestinal mucosal dysfunction with an increased translocation of LPS from gram negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology of depression [no. 1]. Neuroendocrinology Letters 29: 117–124. [PubMed] [Google Scholar]
- 100.Maes M, Kubera M, Leunis J-C, Berk M (2012): Increased IgA and IgM responses against gut commensals in chronic depression: further evidence for increased bacterial translocation or leaky gut [no. 1]. Journal of affective disorders 141: 55–62. [DOI] [PubMed] [Google Scholar]
- 101.Kılıç F, Işık Ü, Demirdaş A, Doğuç DK, Bozkurt M (2020): Serum zonulin and claudin-5 levels in patients with bipolar disorder. Journal of affective disorders 266: 37–42. [DOI] [PubMed] [Google Scholar]
- 102.Ohlsson L, Gustafsson A, Lavant E, Suneson K, Brundin L, Westrin Å, et al. (2019): Leaky gut biomarkers in depression and suicidal behavior. Acta Psychiatrica Scandinavica 139: 185–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Safadi JM, Quinton AM, Lennox BR, Burnet PW, Minichino A (2022): Gut dysbiosis in severe mental illness and chronic fatigue: a novel trans-diagnostic construct? A systematic review and meta-analysis. Molecular Psychiatry 27: 141–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Madison A, Andridge R, Padin A, Wilson S, Bailey MT, Alfano CM, et al. (in press): Endotoxemia Coupled with Heightened Inflammation Predicts Future Depressive Symptoms. Psychoneuroendocrinology. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Wauters L, Van Oudenhove L, Accarie A, Geboers K, Geysen H, Toth J, et al. (2022): Lactobacillus rhamnosus CNCM I-3690 decreases subjective academic stress in healthy adults: a randomized placebo-controlled trial. Gut microbes 14: 2031695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Kelly JR, Allen AP, Temko A, Hutch W, Kennedy PJ, Farid N, et al. (2017): Lost in translation? The potential psychobiotic Lactobacillus rhamnosus (JB-1) fails to modulate stress or cognitive performance in healthy male subjects. Brain, behavior, and immunity 61: 50–59. [DOI] [PubMed] [Google Scholar]
- 107.Cryan JF, O’Riordan KJ, Cowan CS, Sandhu KV, Bastiaanssen TF, Boehme M, et al. (2019): The microbiota-gut-brain axis. Physiological reviews. [DOI] [PubMed] [Google Scholar]
- 108.Lloyd-Price J, Abu-Ali G, Huttenhower C (2016): The healthy human microbiome. Genome medicine 8: 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Joice R, Yasuda K, Shafquat A, Morgan XC, Huttenhower C (2014): Determining microbial products and identifying molecular targets in the human microbiome. Cell metabolism 20: 731–741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Miller R, Kirschbaum C (2018): Cultures under stress: a cross-national meta-analysis of cortisol responses to the Trier Social Stress Test and their association with anxiety-related value orientations and internalizing mental disorders. Psychoneuroendocrinology. [DOI] [PubMed] [Google Scholar]
- 111.Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, et al. (2010): A human gut microbial gene catalogue established by metagenomic sequencing. nature 464: 59–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.