Invited Review
Mood and gut feelings

https://doi.org/10.1016/j.bbi.2009.05.058Get rights and content

Abstract

Evidence is accumulating to suggest that gut microbes (microbiota) may be involved in neural development and function, both peripherally in the enteric nervous system and centrally in the brain. There is an increasing and intense current interest in the role that gut bacteria play in maintaining the health of the host. Altogether the mass of intestinal bacteria represents a virtual inner organ with 100 times the total genetic material contained in all the cells in the human body. Surprisingly, the characterization of this extraordinarily diverse population is only just beginning, since some 60% of these microbes have never been cultured. Commensal organisms live in a state of harmonious symbiosis with each other and their host, however, a disordered balance amongst gut microbes is now thought to be an associated or even causal factor for chronic medical conditions as varied as obesity and inflammatory bowel diseases. While evidence is still limited in psychiatric illnesses, there are rapidly coalescing clusters of evidence which point to the possibility that variations in the composition of gut microbes may be associated with changes in the normal functioning of the nervous system. This review focuses on these data and suggests that the concept should be explored further to increase our understanding of mood disorders, and possibly even uncover missing links to a number of co-morbid medical diseases.

Introduction

Psychiatric disorders which are on the increase globally, already rank among the leading causes of disability, and are expected to take over first place within the next few years. Indeed, the World Health Report 2001 cites depression as causing the largest amount of disability worldwide (disability adjusted life years-DALYs) and in 2004 Ustun et al. stated that depression was the fourth leading cause of disease burden but represents the largest amount of non-fatal burden globally (WHO, 2001, Ustun et al., 2004). In addition, there is extensive epidemiological evidence to support the view that significant co-morbidity exists between many chronic medical and psychiatric diseases, especially mood disorders (Moussavi et al., 2007, Van Lieshout et al., 2008). The severity and prognosis of medical illness are substantially affected by the presence or absence of co-morbid depression. For example, depression is a significant risk factor for myocardial infarction (Rosengren et al., 2004) and its presence at the time of infarction predicts a greater than threefold increase in likely death from cardiac causes within 5 years (Lesperance et al., 2002). Individuals with depression also have higher rates of obesity, hypertension, dyslipidemia, metabolic syndrome and diabetes than the general population (Chengappa et al., 2004, Heiskanen et al., 2006).

Therefore, a better understanding of the biology of mood disorders is critically important not only to provide more effective treatment to patients with these conditions but also to those with chronic co-morbid medical illnesses.

Recent dramatic scientific advances in molecular biology and their application to the study of the human genome have produced much evidence to support the genetic basis of a number of chronic diseases. However, the results are fraught with difficulty of interpretation as well as the knowledge that most of these diseases are polygenic in origin. Indeed the solution to some of the conundra of causation of chronic diseases may lie in greater understanding of the consequences of gene-environment interactions (Cooper, 2003). As a result, the field of epigenetics is expanding explosively and is being applied to psychiatric disorders (Mill and Petronis, 2007, Tsankova et al., 2007).

We believe that one of the most significant areas that need to be investigated in terms of potential environmental factors contributing to both mood disorders and chronic diseases is the external milieu of microbes found in the intestine. In this review we highlight the evidence that is coming together to support the contention that the gut microbiome may be playing a role in neuronal development and plasticity, modulating pain perception and even behavior. This fertile area for exploratory research may shed some light on the causes of, and eventually offer novel therapeutic approaches to, mood disorders and the medical disorders that are often co-morbidly associated with them. (Fig. 1).

Section snippets

The gut microbiome

The human gut is sterile at birth. Immediately after birth, it is colonized by numerous types of microorganisms. In the first weeks of life, tremendous temporal and inter-individual variation is evident in the infant’s microbial populations (microbiome). By 1 year of age, while babies retain their unique bacterial profiles, these converge toward a profile characteristic of the adult individual gastrointestinal tract (Palmer et al., 2007). While significant changes may occur in things such as

Immune system and cytokines

The development of the intestinal immune system is largely dependent upon exposure to microorganisms (Umesaki et al., 1995). In germ free (GF) animals which have scanty lymphoid tissue and constitutively and are almost devoid of immune activity (Talham et al., 1999), association with certain individual selected microorganisms has been shown to be effective in generation of the complete repertoire of immune function. For example, colonization with the segmented filamentous bacterium was able to

Role of gut microbiota in host metabolism

It is widely recognized that gut microbes are responsible for an enormous array of metabolic activities that include effects on the digestion of food and the production of a host of biologically active substances. Recent data suggest that gut microbiota also affect host metabolism, have an impact on energy storage and may therefore be an important environmental factor in the development of obesity and type 2 diabetes (Turnbaugh et al., 2006). These observations are relevant to the

Microbiota and the nervous system

While the role of gut microbiota in influencing brain and nerve function may not be strikingly obvious at first glance, recent research in several different disciplinary fields suggests that intestinal microbiota may be intimately and constitutively involved in modulation of both central and peripheral nerve function. A good example of this influence can be found in clinical medicine in hepatic failure. Hepatic encephalopathyis a commonly encountered medical condition consequent to hepatic

Possible pathways involved in this signaling

The exact mechanisms whereby gut bacteria-induced local changes in gut epithelium and the enteric nervous system communicate with the brain and possibly alter its function and activity, remain to be elucidated. The underlying pathways are highly complex, and it is unlikely that only one common pathway or series of molecules is involved. Two possible pathways we feel may be implicated, however, are reviewed below.

Microbe cell wall constituents

Many different microorganisms may have constitutive and modulatory effects on neuronal function. Commensal microbiota are also being widely consumed by the general public in the form of probiotics. Ingestion of Saccharomyces boulardii, one of these probiotic organisms, has been shown to alter the distribution of the regulatory calcium binding molecule calbindin in the enteric nervous system of conventional pigs (Kamm et al., 2004). The question thus arises as to any common mechanisms of action

Conclusions and perspectives

It is rapidly becoming apparent that the gut microbiome plays a major role in the development and regulation of metabolic systems such as those governing energy production and fat metabolism, neuroendocrine systems such as the HPA axis and the development and function of the immune system. Moreover, evidence is beginning now to accumulate suggesting that intestinal commensals may also be involved in neural development both peripherally in the enteric nervous system and centrally in the brain,

Acknowledgment

We gratefully acknowledge the contribution of the Giovanni and Concetta Guglietti Foundation to some of the research conducted at the Brain-Body Institute.

References (96)

  • B.L. Hart

    Biological basis of the behavior of sick animals

    Neurosci. Biobehav. Rev.

    (1988)
  • F. Holsboer

    Stress, hypercortisolism and corticosteroid receptors in depression: implications for therapy

    J. Affect. Disord.

    (2001)
  • T. Hoverstad et al.

    Short-chain fatty acids in germfree mice and rats

    J. Nutr.

    (1986)
  • L.M. Iyer et al.

    Evolution of cell-cell signaling in animals: did late horizontal gene transfer from bacteria have a role?

    Trends Genet.

    (2004)
  • P.E. Kankaanpaa et al.

    Influence of probiotic supplemented infant formula on composition of plasma lipids in atopic infants

    J. Nutr. Biochem.

    (2002)
  • W. Li et al.

    Memory and learning behavior in mice is temporally associated with diet-induced alterations in gut bacteria

    Physiol. Behav.

    (2009)
  • M. Lyte et al.

    Induction of anxiety-like behavior in mice during the initial stages of infection with the agent of murine colonic hyperplasia Citrobacter rodentium

    Physiol. Behav.

    (2006)
  • D.F. MacFabe et al.

    Neurobiological effects of intraventricular propionic acid in rats: possible role of short chain fatty acids on the pathogenesis and characteristics of autism spectrum disorders

    Behav. Brain Res.

    (2007)
  • S. Moussavi et al.

    Depression, chronic diseases, and decrements in health: results from the World Health Surveys

    Lancet

    (2007)
  • L. O’Mahony et al.

    Lactobacillus and bifidobacterium in irritable bowel syndrome: symptom responses and relationship to cytokine profiles

    Gastroenterology

    (2005)
  • N. Quan et al.

    Brain-immune communication pathways

    Brain Behav. Immun.

    (2007)
  • A. Rosengren et al.

    Association of psychosocial risk factors with risk of acute myocardial infarction in 11119 cases and 13648 controls from 52 countries (the INTERHEART study): case control study

    Lancet

    (2004)
  • R. Schicho et al.

    Hydrogen sulfide is a novel prosecretory neuromodulator in the Guinea-pig and human colon

    Gastroenterology

    (2006)
  • F.A. Schroeder et al.

    Antidepressant-like effects of the histone deacetylase inhibitor, sodium butyrate, in the mouse

    Biol. Psychiatry

    (2007)
  • N. Sudo

    Stress and gut microbiota: does postnatal microbial colonization program the hypothalamic–pituitary–adrenal system for stress response?

    Int. Congr. Ser.

    (2006)
  • M. Tanida et al.

    Effects of intraduodenal injection of Lactobacillus johnsonii La1 on renal sympathetic nerve activity and blood pressure in urethane-anesthetized rats

    Neurosci. Lett.

    (2005)
  • V. Taylor et al.

    Increased rates of obesity in first-presentation adults with mood disorders over the course of four-year follow-up

    J. Affect. Disord.

    (2008)
  • D. Wong et al.

    Cytokines, nitric oxide, and cGMP modulate the permeability of an in vitro model of the human blood–brain barrier

    Exp. Neurol.

    (2004)
  • S. Alesci et al.

    Major depression is associated with significant diurnal elevations in plasma interleukin-6 levels, a shift of its circadian rhythm, and loss of physiological complexity in its secretion: clinical implications

    J. Clin. Endocrinol. Metab.

    (2005)
  • F.A. Amaral et al.

    Commensal microbiota is fundamental for the development of inflammatory pain

    Proc. Natl. Acad. Sci. USA

    (2008)
  • H. Anisman et al.

    Cytokines, stress and depressive illness: brain-immune interactions

    Ann. Med.

    (2003)
  • F. Backhed et al.

    The gut microbiota as an environmental factor that regulates fat storage

    Proc. Natl. Acad. Sci. USA

    (2004)
  • R.H. Belmaker et al.

    Major depressive disorder

    N. Engl. J. Med.

    (2008)
  • A. Bomba et al.

    Improvement of the probiotic effect of micro-organisms by their combination with maltodextrins, fructo-oligosaccharides and polyunsaturated fatty acids

    Br. J. Nutr.

    (2002)
  • L. Capuron et al.

    Association of exaggerated HPA axis response to the initial injection of interferon-alpha with development of depression during interferon-alpha therapy

    Am. J. Psychiatry

    (2003)
  • B.J. Carroll et al.

    Pathophysiology of hypercortisolism in depression

    Acta Psychiatr. Scand. Suppl.

    (2007)
  • K.N.R. Chengappa et al.

    The prevalence of the metabolic syndrome in patients with schizoaffective disorder-bipolar subtype

    Bipolar Disord.

    (2004)
  • M.B. Clarke et al.

    The QseC sensor kinase: a bacterial adrenergic receptor

    Proc. Natl. Acad. Sci. USA

    (2006)
  • R.S. Cooper

    Gene-environment interactions and the etiology of common complex disease

    Ann. Intern. Med.

    (2003)
  • R. Dantzer et al.

    From inflammation to sickness and depression: when the immune system subjugates the brain

    Nat. Rev. Neurosci.

    (2008)
  • S.C. Duncker et al.

    The d-alanine content of lipoteichoic acid is crucial for Lactobacillus plantarum-mediated protection from visceral pain perception in a rat colorectal distension model

    Neurogastroenterol. Motil.

    (2008)
  • A. Fagiolini et al.

    Obesity as a correlate of outcome in patients with bipolar I disorder

    Am. J. Psychiatry

    (2003)
  • B.A. Fallon et al.

    Lyme disease: a neuropsychiatric illness

    Am. J. Psychiatry

    (1994)
  • P. Forsythe et al.

    Oral treatment with live Lactobacillus reuteri inhibits the allergic airway response in mice

    Am. J. Respir. Crit. Care Med.

    (2007)
  • D.N. Frank et al.

    Gastrointestinal microbiology enters the metagenomics era

    Curr. Opin. Gastroenterol.

    (2008)
  • M.P. Freeman et al.

    Omega-3 fatty acids: evidence basis for treatment and future research in psychiatry

    J. Clin. Psychiatry

    (2006)
  • G.M. Gilad et al.

    Overview of the brain polyamine-stress-response: regulation, development, and modulation by lithium and role in cell survival

    Cell. Mol. Neurobiol.

    (2003)
  • C. Grangette et al.

    Enhanced antiinflammatory capacity of a Lactobacillus plantarum mutant synthesizing modified teichoic acids

    Proc. Natl. Acad. Sci. USA

    (2005)
  • Cited by (0)

    View full text