Hostname: page-component-745bb68f8f-d8cs5 Total loading time: 0 Render date: 2025-02-09T14:18:06.884Z Has data issue: false hasContentIssue false

Microbiota-gut-brain research: A critical analysis

Published online by Cambridge University Press:  12 September 2018

Katarzyna B. Hooks
Affiliation:
University of Bordeaux, CBiB, Bordeaux 33076, France. katarzyna.hooks@u-bordeaux.frhttp://kbhooks.wordpress.com/ University of Bordeaux, CNRS/LaBRI, Talence 33405, France
Jan Pieter Konsman
Affiliation:
University of Bordeaux, CNRS/INCIA, Bordeaux 33076, France. jan-pieter.konsman@u-bordeaux.fr
Maureen A. O'Malley
Affiliation:
University of Bordeaux, CNRS/LaBRI, Talence 33405, France University of Bordeaux, CNRS/LaBRI, Talence 33405, France; University of Sydney, School of History and Philosophy of Science, New South Wales 2006, Australiamaureen.omalley@sydney.edu.auhttp://www.maureenomalley.org/
Rights & Permissions [Opens in a new window]

Abstract

Microbiota-gut-brain (MGB) research is a fast-growing field of inquiry with important implications for how human brain function and behaviour are understood. Researchers manipulate gut microbes (“microbiota”) to reveal connections between intestinal microbiota and normal brain functions (e.g., cognition, emotion, and memory) or pathological states (e.g., anxiety, mood disorders, and neural developmental disorders such as autism). Many claims are made about causal relationships between gut microbiota and human behaviour. By uncovering these relationships, MGB research aims to offer new explanations of mental health and potential avenues of treatment.

So far, limited evaluation has been made of MGB's methods and its core experimental findings, many of which are extensively reiterated in copious reviews of the field. These factors, plus the self-help potential of MGB, have combined to encourage uncritical public uptake of MGB discoveries. Both social and professional media focus on the potential for dietary intervention in mental health, and causal relationships are assumed to be established.

Our target article has two main aims. One is to examine critically the core practices and findings of experimental MGB research and to raise questions about them for brain and behavioural scientists who may not be familiar with the field. The other is to challenge the way in which MGB findings are presented. Our positive goal is to suggest how current problems and weaknesses may be addressed, in order for both scientific and public audiences to gain a clearer picture of MGB research and its strengths and limitations.

Type
Target Article
Copyright
Copyright © Cambridge University Press 2019 

1. Introduction

A growing body of “microbiome” research is investigating microbially mediated connections between the gut and brain. Microbes in the gut (“microbiota”) apparently have effects on how humans think, perceive, and experience the world. Numerous scientific articles stress how this research is “revolutionary” and “paradigm-shifting” (e.g., Liu Reference Liu2017; Mayer et al. Reference Mayer, Knight, Mazmanian, Cryan and Tillisch2014). Although such hyperbole is characteristic of microbiome research more generally, many basic views about human capacities are challenged by suggestions that gut microbiota are causally influencing brains and behaviour.

Microbiota-gut-brain (MGB) researchers seek to explain and treat behavioural, cognitive, and mood disorders in host organisms, including humans. The basic methodology is to alter the gut microbiota in rodents, or compare the behaviour of animals with and without microbiota. Some interpretations of the findings from such studies make quite radical claims about the nature of our relationship with our microorganisms and the extent of their control over us. These interpretations propose new ways in which common psychiatric and psychological disorders can be treated, and even normal cognition enhanced. Not surprisingly, these sorts of claims about microbiota and gut-brain connections are of broad interest and have received a great deal of attention in the wider public sphere. Although a critical literature is beginning to develop on both microbiome research generally (Bik Reference Bik2016; Hanage Reference Hanage2014; Quigley Reference Quigley2017) and MGB research in particular (e.g., Bruce-Keller et al. Reference Bruce-Keller, Salbaum and Berthoud2018; Forsythe et al. Reference Forsythe, Kunze and Bienenstock2016), a systematic scrutiny from outside of the field has yet to be conducted.

Our aim is to investigate MGB claims and the research that lies behind them. To do this, we focus on the field's 25 most cited experimental papers of the last decade. We analyse first the methodologies underpinning these core studies, and then their findings, before contextualizing these papers within the wider MGB literature. Our conclusions are cautionary and have a constructive aim. Despite the rapidly increasing body of work in the MGB area, and the wide audiences it reaches, even the most cited papers are at best suggestive. Both methodological and interpretative aspects of this research require consolidation and greater depth. We discuss this message and its broader implications for brain and behavioural research, as well as its communication to a wider audience.

2. Context and historical background

MGB research weaves together several strands of earlier investigation from neuroscience, gastroenterology, and microbiology. The exploration of connections between the gut and brain has a particularly long and venerable research history. Early psychologists William James and Carl Lange are seen as forerunners of brain-gut-axis research (e.g., Eisenstein Reference Eisenstein2016), although they made limited claims about these connections. James merely insisted that “visceral stirrings” should be conceptualized as part of the emotion of fear (1884). Subsequent research continued to connect emotional responses to visceral signals. In the early twentieth century, for example, Walter Bradford Cannon observed that “the movements of the stomach immediately stopped” when “a female [cat] with kittens turned from her state of quiet contentment to one of apparent restlessness” (Cannon Reference Cannon1909, p. 484). He postulated that these changes depended on the sympathetic nerve supply (Cannon Reference Cannon1911).

More fine-grained studies followed. The administration of adrenaline, which is released by the host after activation of the sympathetic nervous system, was discovered to lower the number of pathogenic bacteria needed to establish a generalized infection or to kill the animal (Evans et al. Reference Evans, Miles and Niven1948; Renaud & Miget Reference Renaud and Miget1930). Although these effects were attributed to decreased recruitment of white blood cells (Evans et al. Reference Evans, Miles and Niven1948), it was realized much later that adrenaline also diminishes the bactericidal activity of these cells (Qualliotine et al. Reference Qualliotine, DeChatelet, McCall and Cooper1972). Other experiments revealed that adrenaline actually reduces host mortality after the injection of bacterial toxins (Chedid & Boyer Reference Chedid and Boyer1953; Hodoval et al. Reference Hodoval, Morris, Crawley and Beisel1968), which suggested that this hormone has different effects on living bacteria and bacterial fragments. Another molecule, acetylcholine, which is released by the parasympathetic nervous system of animals, was also shown by other research to be produced by a strain of the bacterium Lactobacillus plantarum (Stephenson & Rowatt Reference Stephenson and Rowatt1947).

By the 1980s, the term brain-gut axis had become a common label for investigations of these connections (e.g., Aziz & Thompson Reference Aziz and Thompson1998; Gastroenterology 1980). A variety of important findings emerged about gut microbes, their cell wall components, and nervous systems or behavioural states (Bluthé et al. Reference Bluthé, Dantzer and Kelley1992; Hart Reference Hart1988; Lyte Reference Lyte1993). Further extensive experimentation on adrenaline and noradrenaline showed that they stimulate the growth of some bacteria (Lyte & Ernst Reference Lyte and Ernst1992) and that some microorganisms are themselves able to produce these substances (Asano et al. Reference Asano, Hiramoto, Nishino, Aiba, Kimura, Yoshihara, Koga and Sudo2012; Tsavkelova et al. Reference Tsavkelova, Botvinko, Kudrin and Oleskin2000). This body of evidence that microbes can influence the gut-brain axis, and in turn be influenced by the brain-gut axis, forms an important basis for more recent developments in MGB research.

From the microbiological angle, intestinal microbes have long been studied for their effects on human health, from the perspectives of both individual pathogens and more systemic community effects (see Haenel Reference Haenel1961; Savage Reference Savage2001). Although work in the 1980s had begun to examine mechanistically how specific intestinal microorganisms might affect mammalian brain states (e.g., Brown et al. Reference Brown, Price, King and Husband1990; Jeppsson et al. Reference Jeppsson, James, Hummel, Brenner, West and Fischer1983), it is only in the last decade that brain-gut-axis research has been able to take advantage of methods that reveal the full diversity of microorganisms inhabiting the human gut. This expanded capacity for the molecular analysis of microbial communities in host organisms is what is now called microbiome research.

Microbiome research developed on the basis of tools that allow analysis of the DNA sequence of entire microbial communities (microbiota). The DNA is directly extracted from microbiota in their natural environments (Handelsman et al. Reference Handelsman, Rondon, Brady, Clardy and Goodman1998; see sect. 5 for more detail). “Microbiomes,” the molecular sequences of these communities, are analysed for compositional patterns and their associations with aspects of the environment. In the mid-2000s, microbiome researchers began to focus more closely on the human ecosystem: the human body and its complement of microorganisms, particularly gut microbes (Eckburg et al. Reference Eckburg, Bik, Bernstein, Purdom, Dethlefsen, Sargent, Gill, Nelson and Relman2005). As human microbiome research developed, key researchers began to use germ-free (GF) mice. These are mice that are born and live their lives without microorganisms until they are experimentally colonized; other GF organisms have been used historically for different purposes (Kirk Reference Kirk2012). Influential studies showed that giving GF mice microbiota transplants from obese hosts could bring about obesity (e.g., Turnbaugh et al. Reference Turnbaugh, Bäckhed, Fulton and Gordon2008). Although GF mice have many abnormalities (see sect. 7.3), they have become the gold experimental standard for causal claims in human microbiome research, which now includes gut-brain studies.

Despite all of these well-known MGB precursors, the current phase of microbiome-oriented gut-brain research often cites its starting point as 2004, when Sudo and colleagues (Reference Sudo, Chida, Aiba, Sonoda, Oyama, Yu, Kubo and Koga2004) used germ-free mice to reveal that “commensal microbes [are] affecting the neural network responsible for controlling stress responsiveness” (p. 271). Many of today's microbiota-gut-brain papers refer to the Sudo et al. paper as “seminal” (e.g., Mayer et al. Reference Mayer, Tillisch and Gupta2015, p. 926; Sampson & Mazmanian Reference Sampson and Mazmanian2015, p. 567) and as a “landmark” in the history of the emerging field of MGB research (e.g., Foster & McVey Neufeld Reference Foster and McVey Neufeld2013, p. 306). This 2004 paper emphasizes a simple potential treatment: probiotics. It also suggests that GF mice allow much of the complexity of microbiomes to be ignored: Mice either have microbiota or they do not. Both this paper and the earlier work have inspired attempts to merge multiple disciplinary perspectives, including those from psychiatry, pharmacology, psychology, neuroscience, immunology, microbiology, and gastroenterology. But in the process of drawing on so many approaches, key problems plaguing broader microbiome analyses were also included: the difficulty of identifying causal pathways and yet the tendency to suggest microbiota are bringing about specific host effects (see Hanage Reference Hanage2014).

3. MGB research and its scope

In part because of its rich historical background, MGB studies draw on a considerable variety of methods and disciplinary approaches (see Supplementary Table 1). These methods are both experimental and descriptive. They focus on implementing microbiota-related interventions that can change specified brain and/or behavioural states. The targets of these interventions are usually disorders of various degrees, including depression (Jiang et al. Reference Jiang, Ling, Zhang, Mao, Ma, Yin, Wang, Tang, Tan, Shi, Li and Ruan2015; Park et al. Reference Park, Collins, Blennerhassett, Ghia, Verdu, Bercik and Collins2013), anxiety (Crumeyrolle-Arias et al. Reference Crumeyrolle-Arias, Jaglin, Bruneau, Vancassel, Cardona, Daugé, Naudon and Rabot2014; Neufeld et al. Reference Neufeld, Kang, Bienenstock and Foster2011a), autism (de Theije et al. Reference de Theije, Wopereis, Ramadan, van Eijndthoven, Lambert, Knol, Garssen, Kraneveld and Oozeer2014; Hsiao et al. Reference Hsiao, McBride, Hsien, Sharon, Hyde, McCue, Codelli, Chow, Reisman, Petrosino, Patterson and Mazmanian2013), schizophrenia (Severance et al. Reference Severance, Yolken and Eaton2016), posttraumatic stress disorder (Hemmings et al. Reference Hemmings, Malan-Müller, van den Heuvel, Demmitt, Stanislawski, Smith, Bohr, Stamper, Hyde, Morton, Marotz, Siebler, Braspenning, Van Criekinge, Hoisington, Brenner, Postolache, McQueen, Krauter, Knight, Seedat and Lowry2017), Parkinson's (Sampson et al. Reference Sampson, Debelius, Thron, Janssen, Shastri, Ilhan, Challis, Schretter, Rocha, Gradinaru, Chesselet, Keshavarzian, Shannon, Krajmalnik-Brown, Wittung-Stafshede, Knight and Mazmanian2016), and anorexia nervosa (Kleiman et al. Reference Kleiman, Watson, Bulik-Sullivan, Huh, Tarantino, Bulik and Carroll2015). But more general brain and behavioural states are also scrutinized, including fear (Bravo et al. Reference Bravo, Forsythe, Chew, Escaravage, Savignac, Dinan, Bienenstock and Cryan2011), stress (O'Mahony et al. Reference O'Mahony, Clarke, Dinan and Cryan2017), mood (Steenbergen et al. Reference Steenbergen, Sellaro, van Hemert, Bosch and Colzato2015), temperament (Christian et al. Reference Christian, Galley, Hade, Schoppe-Sullivan, Kamp Dush and Bailey2015), cognition (Magnusson et al. Reference Magnusson, Hauck, Jeffrey, Elias, Humphrey, Nath, Perrone and Bermudez2015), memory (Gareau et al. Reference Gareau, Wine, Rodrigues, Cho, Whary, Philpott, Macqueen and Sherman2011), and sociability (Desbonnet et al. Reference Desbonnet, Clarke, Shanahan, Dinan and Cryan2014).

When experimental effects are detected, mechanisms are often postulated to consolidate the links made between these brain and behavioural outcomes and the microbiota. Proposed intermediary mechanisms include the vagus nerve, inflammatory molecules, microbial metabolites and “neurotransmitters,” immune system mediators and responses, various “signalling” molecules and cells, the so-called leaky gut, and leaky blood-brain barriers (see Sampson & Mazmanian Reference Sampson and Mazmanian2015). None of these are uncontested as potential or adequate mechanisms. For example, the molecules often labelled “neurotransmitters” are not neurotransmitters for the microbes. Even if these molecules can cross the gut barrier and blood-brain or nerve barriers, they do not meet the criteria for neurotransmitters. These criteria require a neurotransmitter to be present in presynaptic elements, for it to be released in response to presynaptic depolarization and for there to be receptors on a postsynaptic cell (Purves et al. Reference Purves, Augustine, Fitzpatrick, Katz, LaMantia, McNamara and Williams2001). Another very problematic mechanism is the “leaky gut” and its highly disputed role in neurological disorders (e.g., Quigley Reference Quigley2016; Rao & Gershon Reference Rao and Gershon2016; see sect. 7).

An outline of some key studies in MGB research will help show the field's scope and trajectory of development. The now classic Sudo et al. (Reference Sudo, Chida, Aiba, Sonoda, Oyama, Yu, Kubo and Koga2004) paper serves as something of a template for much subsequent research. In that paper, Sudo et al. compare hypothalamo-pituitary-axis (HPA) responses to restraint stress in GF, specific pathogen-free (SPF), and conventional mice (i.e., unmanipulated microbiota). The study found that GF mice show higher post-stress corticosterone concentrations than SPF and conventional mice. In addition, higher corticosterone in GF mice was counteracted by the administration of probiotic bacteria (Bifidobacterium infantis). Because this occurred only to the 9-week-old mice and not the older ones (17 weeks), Sudo et al. (Reference Sudo, Chida, Aiba, Sonoda, Oyama, Yu, Kubo and Koga2004) postulated a crucial developmental stage for the HPA stress response that is determined by microbiota. These key findings of probiotic effects on physiology and behaviour, plus a developmental window of maximum effect, get taken up in numerous other MGB papers.

In 2009, O'Mahony and colleagues established that several consequences of maternal separation stress exist at adulthood: namely, visceral hypersensitivity, changes in gut microbiota, less exploration of novel environments, and more defecation. Those behaviours are often considered “anxiety-like” (see sect. 6 for further discussion). The relationship of such behaviour to microbes had already been explored in earlier work focused on single microbes (e.g., Lyte et al. Reference Lyte, Varcoe and Bailey1998). Following the new trend of focusing more broadly on the microbiota as a whole, Diaz Heijtz et al. (Reference Diaz Heijtz, Wang, Anuar, Qian, Björkholm, Samuelsson, Hibberd, Forssberg and Pettersson2011) and Neufeld et al. (Reference Neufeld, Kang, Bienenstock and Foster2011a) found that GF mice (i.e., no microbiota at all) display fewer anxiety-like behaviours than SPF mice in the light-dark box and elevated plus maze.

In the same year, Bercik et al. (Reference Bercik, Denou, Collins, Jackson, Lu, Jury, Deng, Blennerhassett, Macri, McCoy, Verdu and Collins2011a) published findings of the effects of oral antibiotics on anxiety-like behaviour in the step-down and light preference tests. Comparisons were made after microbiota transplantations into SPF Balb/C mice (an inbred mouse strain widely used in immunology and considered to display a high level of anxiety-like behaviour, or “timidity”), National Institutes of Health (NIH) Swiss mice (an outbred strain that shows less anxiety-like behaviour, or greater “boldness”), or GF Balb/C mice. The study found that oral antibiotic treatment reduced anxiety-like behaviour and increased exploration of the behavioural devices used, and that this increased exploration did not involve autonomic nerves. In addition, Bercik et al. (Reference Bercik, Denou, Collins, Jackson, Lu, Jury, Deng, Blennerhassett, Macri, McCoy, Verdu and Collins2011a) reported that Balb/C recipient mice transplanted with NIH Swiss microbiota showed more exploration than their counterparts with only Balb/C microbiota. Conversely, NIH Swiss mice that received Balb/C microbiota transplantation displayed less exploration than those that were colonized with NIH Swiss microbiota. The success of these interventions suggested to many people in the field that the microbiota is a major causal agent in determining anxiety-like behaviour.

Making a narrower microbial intervention (i.e., just one microbe, not a community), Bravo et al. (Reference Bravo, Forsythe, Chew, Escaravage, Savignac, Dinan, Bienenstock and Cryan2011) used a probiotic bacterium (Lactobacillus) to manipulate anxiety-like and depression-related behaviours in mice. They examined depression-related behaviour with the forced swim test (measuring how long the animal was immobile) and anxiety-related behaviour by the number of entries on to the open arms of the elevated plus maze. They also measured the time spent freezing after fear conditioning with a mild electric shock. Probiotic administration reduced immobility during forced swim tests and increased the number of open arm entries in the elevated maze. Subdiaphragmatic vagotomy (severing the vagus nerve under the diaphragm) prevented these effects (however, see Bercik et al. [Reference Bercik, Denou, Collins, Jackson, Lu, Jury, Deng, Blennerhassett, Macri, McCoy, Verdu and Collins2011a], who found no role for the vagus nerve in modulating the effects of antibiotics on the behaviour of mice in the light-dark preference and step-down tests). Follow-up studies subsequently showed that the probiotic facilitates firing of vagal sensory fibres (Perez-Burgos et al. Reference Perez-Burgos, Wang, Mao, Mistry, McVey Neufeld, Bienenstock and Kunze2013). Findings such as these have given rise to the idea of “psychobiotics.” These are substances derived from microorganisms that can be used as treatments for improving mental health (Dinan et al. Reference Dinan, Stanton and Cryan2013). This notion has strong appeal inside and outside of the MGB field.

A study by Hsaio et al. (Reference Hsiao, McBride, Hsien, Sharon, Hyde, McCue, Codelli, Chow, Reisman, Petrosino, Patterson and Mazmanian2013) suggested how such interventions might work mechanistically. The authors used adult mice born from mothers that had been administered an immune stimulation (a viral mimic) during pregnancy. The pups were born with both a “leaky gut” and the behavioural features of autistic developmental disorders. The adult offspring displayed anxiety-like features in the open field, stereotypical behaviour, less social interaction, and fewer ultrasound vocalizations. Feeding Bacteroides fragilis to the impaired mice mitigated “obsessive” behaviours such as grooming and marble-burying. However, reduced sociability did not improve, which was attributed to developmental timing. B. fragilis was known from earlier immunological studies to improve immune defects (Mazmanian et al. Reference Mazmanian, Round and Kasper2008). Although Hsaio et al. did not isolate colonized B. fragilis in the mouse intestines, a metabolic mediator associated with this microorganism was restored to normal levels after probiotic treatment. Studies such as this, although still incomplete, hint at the potential mechanistic pathways that might underlie microbiota effects on brain and behaviour.

Many MGB studies, including those just discussed, are believed to be relevant to human psychiatric disorders. In addition, cognitive and behavioural processes that are not necessarily connected to any psychiatric disorder have also been linked to microbiota changes. Bravo et al. (Reference Bravo, Forsythe, Chew, Escaravage, Savignac, Dinan, Bienenstock and Cryan2011) showed that although no differences in the amount of behavioural freezing were observed immediately after mice received a foot shock, mice that were fed a probiotic showed more conditioned freezing the next day than probiotic-free mice. Diet also has effects. Non-obese antibiotic-pretreated mice were given microbiota transplants from animals fed a high-fat diet. The mice with the high-fat microbiota transplants displayed more conditioned freezing to a shock-signalling tone than did mice with transplants from animals on a control diet (Bruce-Keller et al. Reference Bruce-Keller, Salbaum, Luo, Blanchard, Taylor, Welsh and Berthoud2015). Gareau et al. (Reference Gareau, Wine, Rodrigues, Cho, Whary, Philpott, Macqueen and Sherman2011) observed that probiotics could reverse stress-induced deficits in novel object recognition. Antibiotic treatment of healthy mice from adolescence through adulthood was also found to impair novel object recognition in mice (Desbonnet et al. Reference Desbonnet, Clarke, Traplin, O'Sullivan, Crispie, Moloney, Cotter, Dinan and Cryan2015).

Whether about cognitive or emotional capacities, or aspects of psychiatric disorders, the potential implications of these and many other studies are striking. Many of the core findings and interpretations are echoed repeatedly in the general MGB literature, which is characterized by an abundance of reviews (see sect. 1 in the Supplementary Material). Some of this work then goes so far as to claim that microbes control the mind and that free will is thereby refuted (e.g., Lepage et al. Reference Lepage, Leclerc, Joossens, Mondot, Blottière, Raes, Ehrlich and Doré2013; Stilling et al. Reference Stilling, Dinan and Cryan2016; see sect. 7). Most of these reviews, as well as much primary research, proclaim that a conceptual and methodological revolution is underway in brain and behavioural research (e.g., Liu Reference Liu2017; Mayer et al. Reference Mayer, Knight, Mazmanian, Cryan and Tillisch2014). And yet much of the research is highly speculative regarding causation and mechanisms, some of it is contradictory, and many well-established methods are used in limited, mistaken, and even outdated ways, as we will show.

Although some scientific papers and popular essays have already pointed toward central problems for microbiome research (e.g., Eisen Reference Eisen2017; Shanahan & Quigley Reference Shanahan and Quigley2014) and warnings have been issued about MGB “hype” in particular (see comments in Smith Reference Smith2015; Zimmer Reference Zimmer2014), these discussions have not been based on detailed examinations of the core literature. Very commonly within the field, cautionary statements are embedded in strongly promotional overviews of MGB research (e.g., Mayer et al. Reference Mayer, Knight, Mazmanian, Cryan and Tillisch2014; Sherwin et al. Reference Sherwin, Dinan and Cryan2018). Our aim is to provide a more thorough critical and external analysis of the field for anyone who wants to understand human minds and behaviours, and their putative microbiome connections.

4. The 25 most cited MGB papers

In order to analyse the field more closely, we examined the most highly cited MGB papers in the last decade (Table 1). We chose this set of papers because of their importance to the established field, and particularly its experimental core. They have shaped the field and continue to structure it, as all their citations attest. Focusing on them allows us to probe deeply into influential methods and interpretations, which would be less effectively achieved in a comprehensive but relatively shallow overview of all existing literature. Although we recognize that this selection of papers will not include the most recent work in the field (some of which may be using improved techniques), our aim here is to capture the most recognized experimental work that has been the basis for the majority of reviews and subsequent studies, as well as media attention.

Table 1. The 25 most cited papers in MGB researcha

a Papers were extracted using a combination of PubMed searches and Google Scholar citations. See the main text for detailed selection methods. Papers are ranked by the number of citations received.

To identify this central corpus of work, we carried out a PubMed search using the term “gut-brain microbiota” (date of access: May 25, 2017; updated July 11, 2018). We discarded all reviews, which formed a very high proportion of the literature (almost 50%; see Supplementary Material). This search found 325 articles. We then used Google Scholar citation counts for each article to rank all the papers with more than 150 citations (a total of 15). To supplement this core of highly cited papers, we also examined the references to open access articles within the original 325 articles. This strategy found another 9 highly cited articles. Finally, we conducted a third search using the looser term “brain microbiota.” This search found 867 articles. We inspected the most cited articles of this group, which revealed another 3 publications that had not appeared in our earlier “gut-brain microbiota” search. We slightly cropped this list to 25 papers, of which the lowest number of citations is just over 120 and the highest above 1300 (Table 1). We then analysed the text of these papers manually, with an initial focus on two categories of methodology: microbiome methods (sect. 5) and behavioural tests and statistics (sect. 6).

5. Microbiome methodology

Microbiome research relies on the rapid and extensive DNA profiling of bacterial and other microorganismal genomes in specified locations. This use of DNA sequencing tools to explore microbial biodiversity is often called metagenomics, meaning that it goes beyond the single-species genome analyses of genomics (Handelsman Reference Handelsman2004). It allows the investigation of microbial communities in a vast variety of environments, including those provided by animal hosts. These methods have liberated the study of microbial biodiversity from the constraints of pure culture. Pure culturing approaches require growing microorganisms in the laboratory, which is not feasible (yet) for many microorganismal groups.

In the simplest scenario for sequencing, the presence of species is evaluated with metagenomic methods, which can be performed in two ways. The first is tag (or amplicon) sequencing, usually of a particular stretch of a ribosomal gene. The second is shotgun sequencing, which captures all the genes in the environmental sample. Tag sequencing is still widely used despite being restricted to information about bacterial abundance and diversity. Shotgun sequencing provides more information about the total pool of genes present in the environment but requires more complicated bioinformatic analysis. In order to do more than catalogue taxa on the basis of genes, researchers also employ metatranscriptomic methods to find actively transcribed genes, and metabolomic analyses to quantify the output of bacterial metabolic pathways (see Knight et al. [Reference Knight, Vrbanac, Taylor, Aksenov, Callewaert, Debelius, Gonzalez, Kosciolek, McCall, McDonald, Melnik, Morton, Navas, Quinn, Sanders, Swafford, Thompson, Tripathi, Xu, Zaneveld, Zhu, Caporaso and Dorrestein2018] for an updated methodological primer). However, whether tag or shotgun methods are used, the bulk of microbiome research has yet to advance beyond gene catalogues, and this greatly limits what can be said about microbial effects on hosts and other environments. But as we will show, a surprising amount of the MGB research in our top-cited sample does not even achieve the cataloguing step.

The gut is home to the most studied but also the most complex human-associated microbiota. It contains hundreds if not thousands of different microbial species, of which bacteria are the main component and research focus. The relative abundance and diversity of bacteria can vary considerably from one individual human to another (Human Microbiome Project Consortium 2012). Difficulties in interpreting diverse and complex sequence data result in the main output of health-focused microbiome studies being simple correlations between the abundance of particular taxa and host-associated disease states. These association patterns do not allow cause and effect to be ascertained (de Vos & de Vos Reference de Vos and de Vos2012; Hanage Reference Hanage2014). Moreover, the great majority of investigation is done with faecal samples, which are unlikely to represent microbial activity in the gut itself, especially in the small intestine or in association with the mucosal surface (Gevers et al. Reference Gevers, Kugathasan, Denson, Vázquez-Baeza, Van Treuren, Ren, Schwager, Knights, Song, Yassour, Morgan, Kostic, Luo, González, McDonald, Haberman, Walters, Baker, Rosh, Stephens, Heyman, Markowitz, Baldassano, Griffiths, Sylvester, Mack, Kim, Crandall, Hyams, Huttenhower, Knight and Xavier2014; Momozawa et al. Reference Momozawa, Deffontaine, Louis and Medrano2011; Quigley Reference Quigley2017). Nevertheless, the sheer convenience of such samples continues to ensure their popularity.

How does microbiome research feature in MGB studies? In general, most MGB papers are not microbiome-driven in the way many other health-related or environmental microbiome papers are. In fact, in MGB research, including our 25 most cited list, “microbiota” and “microbiome” are often used simply to indicate that microorganisms in the human body appear to be involved in producing observed effects. Despite many methodological advances in microbiome research, standard microbiome analyses are not carried out even in many of the most highly cited MGB papers.

There are four broad categories of “microbiota” methods in the 25 most cited MGB papers we analysed.

  1. 1. Comparisons of behaviours in GF mice/rat microbiomes with conventionally colonized or SPF animals (e.g., Crumeyrolle-Arias et al. Reference Crumeyrolle-Arias, Jaglin, Bruneau, Vancassel, Cardona, Daugé, Naudon and Rabot2014; Gareau et al. Reference Gareau, Wine, Rodrigues, Cho, Whary, Philpott, Macqueen and Sherman2011; Sudo et al. Reference Sudo, Chida, Aiba, Sonoda, Oyama, Yu, Kubo and Koga2004). Sometimes a rescue experiment is performed in which a standard microbiota is transplanted into GF animals to investigate whether the phenotype can be reversed (Clarke et al. Reference Clarke, Grenham, Scully, Fitzgerald, Moloney, Shanahan, Dinan and Cryan2013; Diaz Heijtz et al. Reference Diaz Heijtz, Wang, Anuar, Qian, Björkholm, Samuelsson, Hibberd, Forssberg and Pettersson2011; Neufeld et al. Reference Neufeld, Kang, Bienenstock and Foster2011a; Reference Neufeld, Kang, Bienenstock and Foster2011b).

  2. 2. Studies of normally colonized mice treated with antibiotics (Ait-Belgnaoui et al. Reference Ait-Belgnaoui, Durand, Cartier, Chaumaz, Eutamene, Ferrier, Houdeau, Fioramonti, Bueno and Theodorou2012; Bajaj et al. Reference Bajaj, Heuman, Sanyal, Hylemon, Sterling, Stravitz, Fuchs, Ridlon, Daita, Monteith, Noble, White, Fisher, Sikaroodi, Rangwala and Gillevet2013; Bercik et al. Reference Bercik, Denou, Collins, Jackson, Lu, Jury, Deng, Blennerhassett, Macri, McCoy, Verdu and Collins2011a; Desbonnet et al. Reference Desbonnet, Clarke, Traplin, O'Sullivan, Crispie, Moloney, Cotter, Dinan and Cryan2015). One study in our sample then re-colonized the animals with microbiota from obese and normal hosts (Bruce-Keller et al. Reference Bruce-Keller, Salbaum, Luo, Blanchard, Taylor, Welsh and Berthoud2015).

  3. 3. Studies in which probiotics and placebos are given to human or other animal subjects (Supplementary Table 2).

  4. 4. Standard microbiota studies that assess the experimental alteration of gut microbes (Supplementary Table 3). Some older methods are still used to describe the microbial community, such as denaturing gel electrophoresis (DGGE) or terminal restriction fragment length polymorphism (T-RFLP). But at least some MGB researchers are now turning to more contemporary methods such as quantitative polymerase chain reaction (qPCR), which is an amplification method that targets specific molecules and thus selected taxa, or shotgun DNA sequencing that encompasses the whole community.

For most of the interventions in the third category of “microbiota” methods (probiotics), Bifidobacterium sp. and Lactobacillus sp. are the probiotics of choice, with Lactobacillus helveticus being the most popular (Supplementary Table 2). These genera of organisms have long been traditional targets for claims about fermented milk products having digestive and physiological benefits (e.g., Metchnikoff Reference Metchnikoff1908). B. fragilis, the intervention microorganism in Hsiao et al.’s (Reference Hsiao, McBride, Hsien, Sharon, Hyde, McCue, Codelli, Chow, Reisman, Petrosino, Patterson and Mazmanian2013) study, is not found in fermented milk products but can be deployed according to the World Health Organization (WHO) definition of a probiotic: any live microorganism that is used to intervene in a human body to bring about health effects (Hill et al. Reference Hill, Guarner, Reid, Gibson, Merenstein, Pot, Morelli, Canani, Flint, Salminen, Calder and Sanders2014; however, see Shanahan & Quigley [Reference Shanahan and Quigley2014] for conceptual concerns). We will come back to probiotics and their implications in section 7.

An important observation to make here is that treatment with single or multiple probiotics is not strictly a “microbiota” or “microbiome” study. Normally, this term is reserved for studies in which microbiota samples are analysed bioinformatically after sequencing. In MGB probiotic research, however, researchers might not even profile changes in bacterial composition, and when they do, no differences may be observed (e.g., Tillisch et al. Reference Tillisch, Labus, Kilpatrick, Jiang, Stains, Ebrat, Guyonnet, Legrain-Raspaud, Trotin, Naliboff and Mayer2013). Surprisingly, even when microbiota are analysed for changes, very limited microbiome methodology is used (Supplementary Table 3). The methods that are employed are often not state-of-the-art. It is curious indeed to see much older qualitative methods, such as DGGE, being used for a publication in 2013 (Park et al. Reference Park, Collins, Blennerhassett, Ghia, Verdu, Bercik and Collins2013). Although a useful tool in the 1990s, community fingerprinting methods like DGGE and T-RFLP have long been superseded by more advanced quantitative sequencing methods. These newer methods allow closer analysis of the composition and potential function of microbial communities.

It is important to note, however, that microbiome research in general continues to have a “causality problem” despite improved sequence analysis tools (Hanage Reference Hanage2014). Many microbiome studies simply cannot isolate specific causes no matter how sophisticated their sequencing and bioinformatic tools; even the experimental work with microbiota transplants is not adequate to demonstrate whole-microbiome causality (O'Malley & Skillings Reference O'Malley and Skillings2018; see sect. 7). In this regard, MGB studies may have an advantage, in that they focus on single microorganisms (probiotics) or small groups of microbes that can be manipulated. However, a probiotic focus would not normally license claims about the whole microbiome, and even narrow probiotic causal claims are problematic (see sect. 7).

A standard interpretation in MGB research is to attribute differences in behaviour between GF and non-GF animals to the lack of microbiota in the former (ditto for antibiotic interventions, which deplete but do not fully remove the microbiota). Often the different treatments experienced by GF or antibiotic-treated mice are not remarked on. Few studies in our most cited sample provide controls that would enable singling out the effects of the microbiota itself (e.g., rescue of phenotype by re-infecting GF animals with a full community transplant or by reintroducing specific bacteria). Although GF models have yielded many interesting results, questions continue to be asked about how relevant they are to humans (Nguyen et al. Reference Nguyen, Vieira-Silva, Liston and Raes2015), because very few humans ever experience germ-free conditions. Although sometimes GF status is equated with environments that have high levels of hygiene and multiple antibiotic treatments (e.g., neonatal care facilities; see Clarke et al. Reference Clarke, Grenham, Scully, Fitzgerald, Moloney, Shanahan, Dinan and Cryan2013), for the majority of researchers these are not considered equivalent conditions at all.

Overall, there are very few studies in this highly cited group of papers that have an experimental approach genuinely able to demonstrate the impact of the microbiota itself on behaviour. Correlations are loosely interpreted as indications of potential mechanisms (however, see Bajaj et al. [Reference Bajaj, Heuman, Sanyal, Hylemon, Sterling, Stravitz, Fuchs, Ridlon, Daita, Monteith, Noble, White, Fisher, Sikaroodi, Rangwala and Gillevet2013] for a more sophisticated analysis of correlation networks of microbial metabolites). The conditions under which potential mechanisms might operate are not specified. For example, one study postulates “the existence of a gut–brain axis in alcohol dependence, in which the gut microbiota could alter the gut-barrier function and influence behavior in alcohol dependence” (Leclercq et al. Reference Leclercq, Matamoros, Cani, Neyrinck, Jamar, Stärkel, Windey, Tremaroli, Bäckhed, Verbeke, de Timary and Delzenne2014, p. E4491). Yet all that this particular piece of research demonstrates is a correlation between increased intestinal permeability and certain bacterial taxa. Less cited and newer studies may be making greater efforts to show microbiota causality of behaviour and brain function (see sect. 8), but, in general, invoking the whole microbiome, rather than specific members of it, will require methods that are carefully designed to deal with the complexities of thousands of interacting organisms and pathways.

One consequence of this complexity is that inter-individual variability between human microbiomes is so high that it is impossible – given most clinical sampling practices – to distinguish specific groups of patients or animals and to find the taxa most associated with different health states (e.g., Falony et al. Reference Falony, Joossens, Vieira-Silva, Wang, Darzi, Faust, Kurilshikov, Bonder, Valles-Colomer, Vandeputte, Tito, Chaffron, Rymenans, Verspecht, De Sutter, Lima-Mendez, D'Hoe, Jonckheere, Homola, Garcia, Tigchelaar, Eeckhaudt, Fu, Henckaerts, Zhernakova, Wijmenga and Raes2016). Frequently, when differences in bacterial composition are observed in the broader body of MGB literature, they are simple correlations from single studies rather than multiple comparative analyses. Considering that hundreds of taxa are involved in any gut community, it is not surprising that some correlations are found. The broader microbiome field (outside MGB) uses a range of statistical correction measures, and their implementation – although still imperfect – at least reduces gross false discovery rates (Knight et al. Reference Knight, Vrbanac, Taylor, Aksenov, Callewaert, Debelius, Gonzalez, Kosciolek, McCall, McDonald, Melnik, Morton, Navas, Quinn, Sanders, Swafford, Thompson, Tripathi, Xu, Zaneveld, Zhu, Caporaso and Dorrestein2018; Weiss et al. Reference Weiss, Van Treuren, Lozupone, Faust, Friedman, Deng, Xia, Xu, Ursell, Alm, Birmingham, Cram, Fuhrman, Raes, Sun, Zhou and Knight2016). For example, one of the reasons that standard parametric tests are not adapted to microbiome data is because of the issue of compositionality. Rapid changes to any single taxon in the microbiota are often measured as changes to all of the taxa, instead of reflecting true abundances. This property leads to extremely high false discovery rates. These ongoing issues add to the field's struggles to achieve causal explanations of phenomena such as disease, but their incidence in MGB research is exacerbated by weaknesses in the methods that are used in combination with microbiome analyses.

6. Neuroendocrine, behavioural, and statistical tests

Microbiome research in its standard sense (i.e., the sequencing and bioinformatic analysis of community genomes) might inform only a subset of MGB papers, and even when it is carried out, it is unlikely to be the methodological focus. Most of the methodology is in fact centred on rodent hormones and behaviour in different conditions. We divided the 25 most cited MGB papers into five categories according to their research focus relative to hormones and behaviour: (1) neuroendocrine “stress” axis; (2) emotion-mood: anxiety; (3) mood disorder: depression; (4) autism spectrum/developmental disorders; and (5) cognition (see Supplementary Tables 4a–4e). About half of the 25 top-cited papers are concerned with activation of the so-called neuroendocrine “stress” axis, which results in the production of stress-related glucocorticoid hormones (Supplementary Table 4a). All of these studies, save one, describe experimental work done in rodents. Sixteen of the top 25 papers explore anxiety, of which 13 studies were carried out on rodents (Supplementary Table 4b). A little less than a quarter (6) of the articles are related to depression, with the majority of that work being done in humans (Supplementary Table 4c). Only two studies present work on animal models of autism spectrum disorder (Supplementary Table 4d), and six address different forms of cognition (Supplementary Table 4e).

Most of the studies we examined do not explicitly justify their methodologies. They seldom address potentially confounding effects (e.g., maternal separation and water avoidance stress) that may complicate interpretation and limit the generalizability of findings. The adequacy of particular behavioural tests and measures is rarely discussed and seems to be taken for granted (admittedly because many other studies have done so). For example, following Sudo et al.’s initial Reference Sudo, Chida, Aiba, Sonoda, Oyama, Yu, Kubo and Koga2004 work, about half of the papers in our top-cited sample measure corticosterone in relation to gut microbiota in rodents. Although most of this subset of papers examines corticosterone in the context of stress – a framework laid down by formative research published 60 years ago (Eik-Nes & Samuels Reference Eik-Nes and Samuels1958; Gold et al. Reference Gold, Singleton, Macfarlane and Moore1958; Persky et al. Reference Persky, Hamburg, Basowitz, Grinker, Sabshin, Korchin, Herz, Board and Heath1958) – it is worth recalling that non-stressful events, such as meal consumption, also increase the circulating concentration of this glucocorticoid (Toda et al. Reference Toda, Morimoto, Nagasawa and Kitamura2004; Wang et al. Reference Wang, Dourmashkin, Yun and Leibowitz1999). Adrenalin can equally be considered a stress hormone (Mormède et al. Reference Mormède, Andanson, Aupérin, Beerda, Guémené, Malmkvist, Manteca, Manteuffel, Prunet, van Reenen, Richard and Veissier2007). In other words, there can be confounding factors at play in any observation of stress responses.

The appropriateness of animal models for human disease is seldom argued for and yet is of crucial importance for the implications of these studies. Not only do mice and humans have different gut structure and neuroanatomy, different microbiota, and different evolved behaviours (see Arrieta et al. Reference Arrieta, Walter and Finlay2016; Nguyen et al. Reference Nguyen, Vieira-Silva, Liston and Raes2015), but there are also acute problems of “translation” into clinical practice when it comes to claims about stress, anxiety, and depression. Behaviours that may be normal for mice (e.g., fearfulness and timidity) are not normal or desirable for humans, and vice versa. Moreover, no self-report-based evaluations can be made on rodents to gain better insight into the organism's experience. Although terminology about findings related to disorders is generally appropriate in the 25 papers we examined most closely (e.g., “anxiety-like” and “depression-like”), we nevertheless found several instances of terms for multidimensional human disorders (e.g., “anxiety” and “depression”) being applied to the unidimensional rodent results (see Supplementary Tables 5a, 5b).

Translational issues arise in any research that extrapolates from rodent models to human function (Zeiss & Johnson Reference Zeiss and Johnson2017) but are particularly pertinent to neuropsychiatric disorders (Homberg Reference Homberg2013). In rodent behavioural studies, interpretations of results obtained in the open field, elevated plus maze, light-dark box, and forced swim tests have frequently been criticized. Indeed, some critical reviews recommend finding new animal paradigms to investigate anxiety (Belzung & Griebel Reference Belzung and Griebel2001). Some authors go so far as to say that “evidence in support of the validity of the plus-maze, the light/dark box and the open-field as anxiety tests is poor and methodologically questionable” (Ennaceur Reference Ennaceur2014, p. 55). Other authors consider increased immobility in the forced swim test an adaptive passive coping strategy rather than a measure of the behavioural despair that is indicative of human depression-like behaviour (Commons et al. Reference Commons, Cholanians, Babb and Ehlinger2017; Molendijk & de Kloet Reference Molendijk and de Kloet2015).

When articles from our 25 most cited papers do take notice of translational issues, they may not take them seriously. For example, Hsiao et al. (Reference Hsiao, McBride, Hsien, Sharon, Hyde, McCue, Codelli, Chow, Reisman, Petrosino, Patterson and Mazmanian2013, p. 1456) quote Bourin et al. (Reference Bourin, Petit-Demoulière, Dhonnchadha and Hascöet2007) as saying that “mapping an animal's movement in an open arena” allows researchers “to measure … anxiety.” Crucially, however, Bourin and colleagues are arguing that it is important to specify whether the open field test is used under dimly lit conditions to measure mere locomotor activity, or whether implementing it in bright light is testing innate rodent anxiety of open spaces during the day. Bourin et al. (contra Hsiao et al.’s interpretation) go on to urge caution about interpreting findings as having implications for anxiety disorders (Bourin et al. Reference Bourin, Petit-Demoulière, Dhonnchadha and Hascöet2007). In the broader MGB field (i.e., beyond the top-cited papers), there are some examples of researchers supplementing or changing their reliance on the open field and elevated maze plus tests (e.g., Bassi et al. Reference Bassi, Kanashiro, Santin, de Souza, Nobre and Coimbra2012; Goehler et al. Reference Goehler, Park, Opitz, Lyte and Gaykema2008), in order to avoid the confounding of anxiety-like behaviour with simple alterations in locomotor activity patterns (Swiergiel & Dunn Reference Swiergiel and Dunn2007). Most commonly, however, if mentioning these issues, MGB researchers merely note them and then very pragmatically continue with animal model manipulations and interpretations.

To conclude our methodological analysis, there are reasons to think that the statistical analyses carried out by some MGB studies in our most cited sample are not appropriate (see Supplementary Table 6). In particular, one-way analyses of variance (ANOVAs) or Student's t-tests are frequently employed when the experimental design includes more than one independent variable. In such cases (e.g., Ait-Belgnaoui et al. Reference Ait-Belgnaoui, Durand, Cartier, Chaumaz, Eutamene, Ferrier, Houdeau, Fioramonti, Bueno and Theodorou2012; Reference Ait-Belgnaoui, Colom, Braniste, Ramalho, Marrot, Cartier, Houdeau, Theodorou and Tompkins2014; Ohland et al. Reference Ohland, Kish, Bell, Thiesen, Hotte, Pankiv and Madsen2013), two- or three-way ANOVAs are required. In many biological situations, the effect of one factor on an outcome of interest often depends on other factors. Hence, when two or more independent variables or factors (such as microbiota status and stress) are studied, it is important to address both the effects of those factors independently and their interaction with the dependent variable being measured (e.g., behaviour in a specific test). Several of the 25 most cited papers did not do this (Supplementary Table 6). Finally, in a few of the MGB papers we analysed, statistically negative results (P > 0.10) are presented as if they are positive findings. For example, non-significant findings after intervention strategies on the microbiota are still used to argue for potential microbiome effects (see Bailey et al. Reference Bailey, Dowd, Galley, Hufnagle, Allen and Lyte2011; Bravo et al. Reference Bravo, Forsythe, Chew, Escaravage, Savignac, Dinan, Bienenstock and Cryan2011; Tillisch et al. Reference Tillisch, Labus, Kilpatrick, Jiang, Stains, Ebrat, Guyonnet, Legrain-Raspaud, Trotin, Naliboff and Mayer2013). It would be much more straightforward to say “no effect is found” without assuming other methods or future experiments on larger cohorts will find the desired outcomes.

Following Fisher, it is standard in the life sciences to consider P < 0.05 as statistically significant and, conversely, that P > 0.05 indicates a non-significant difference (Habibzadeh Reference Habibzadeh2013). In this context, it is not possible to talk about “marginally significant” or “partially significant” (Habibzadeh Reference Habibzadeh2013), or as noted previously, “potentially significant.” At best, a statistical trend can be inferred when 0.10 < P < 0.05, provided there is sufficient statistical power. But if anything, studies in the life sciences tend to be underpowered, which has led several authors to make a plea for the use of more stringent cut-offs for P values and to consider only P < 0.01 as statistically significant (e.g., Colquhoun Reference Colquhoun2014; Vidgen & Yasseri Reference Vidgen and Yasseri2016). MGB research has yet to reflect on this advice.

These behavioural and statistical testing problems are by no means exclusive to MGB research. In fact, they are common throughout rodent-based behavioural neuroscience (Button et al. Reference Button, Ioannidis, Mokrysz, Nosek, Flint, Robinson and Munafò2013). But in MGB research, these weaknesses are compounded by the fact that it is misleading in some of the papers even to refer to microbiomes because no such analysis is done. Even when it is, superseded methods are providing very low-quality analyses. It is difficult of course to do everything well in interdisciplinary research, but, in some instances, it seems as if MGB papers are simply invoking the term “microbiome” without appreciating the minimal methodological commitments with which the term is normally accompanied.

7. Strong claims and interpretations

Although many of our 25 most cited papers use fairly basic reasoning, with limited mechanistic detail, they do not by and large indulge in the overinterpretation and overstatement to the extent we found in some of the broader MGB research literature. However, both our smaller sample of top-cited papers and the larger body of literature we examined divulge many examples of papers in which strong claims – such as “conclusively demonstrate” and “conclusive proof” (e.g., Ait-Belgnaoui et al. Reference Ait-Belgnaoui, Colom, Braniste, Ramalho, Marrot, Cartier, Houdeau, Theodorou and Tompkins2014; Bravo et al. Reference Bravo, Forsythe, Chew, Escaravage, Savignac, Dinan, Bienenstock and Cryan2011) – are offset by more conservative elaborations, sometimes in the very same paper (e.g., Christian et al. Reference Christian, Galley, Hade, Schoppe-Sullivan, Kamp Dush and Bailey2015; Foster & McVey Neufeld Reference Foster and McVey Neufeld2013). We are tempted to diagnose this as a case of “double-dipping,” when cautionary statements are belied by much more dramatic claims. We believe this strategy influences the public uptake of MGB research. In the following sub-sections, we discuss a selection of the overblown conclusions or speculations that help inflame the field, from the most abstract to the highly practical. We do this in order to show how misinterpretation may arise and propagate, especially in the review papers that are so dominant in MGB literature (between 40% and 50%; see sect. 1 in the Supplementary Material).

7.1. Claims about causality and determinism

In the wider field of health-related microbiome research, there are many recognized difficulties in extracting cause-effect relationships from microbiome data (e.g., Hanage Reference Hanage2014; Surana & Kasper Reference Surana and Kasper2017), largely because of how the standard methodology works. Microbiome analysis is basically descriptive, not explanatory. Many efforts are currently underway to explore and assess causal claims, but these attempts are hampered by the whole-community focus of much microbiome methodology. Because microbiome methods begin with communities, there are often expectations that explanations will be found at the community level too, rather than at the level of populations of individual organisms and specific biochemical pathways (e.g., O'Malley & Skillings Reference O'Malley and Skillings2018; Rosen & Palm Reference Rosen and Palm2017).

We can see this problem most clearly when MGB researchers attribute changes in human health to changes in the community of gut microorganisms. These changes can be simple shifts in the relative proportions of groups of microorganisms in the community (e.g., Bailey et al. Reference Bailey, Dowd, Galley, Hufnagle, Allen and Lyte2011; Jiang et al. Reference Jiang, Ling, Zhang, Mao, Ma, Yin, Wang, Tang, Tan, Shi, Li and Ruan2015) or in reference to “normal” community compositions (Clarke et al. Reference Clarke, Grenham, Scully, Fitzgerald, Moloney, Shanahan, Dinan and Cryan2013; Leclercq et al. Reference Leclercq, Matamoros, Cani, Neyrinck, Jamar, Stärkel, Windey, Tremaroli, Bäckhed, Verbeke, de Timary and Delzenne2014). One of our top-25 articles attributed memory-regulating causality to the mere presence of a microbiota, rather than any particular composition (Gareau et al. Reference Gareau, Wine, Rodrigues, Cho, Whary, Philpott, Macqueen and Sherman2011), as did Sudo et al. (Reference Sudo, Chida, Aiba, Sonoda, Oyama, Yu, Kubo and Koga2004) for stress response. This is a general message gleaned from GF mouse studies, where the causal variable can be the simple presence or absence of a microbiota. In other papers, community-level differences are often assigned causal roles under the banner of “dysbiosis.”

Dysbiosis is frequently defined as either a broad change or an “imbalance” in microbiota that produces a diseased state in the (human) host (e.g., Mazmanian et al. Reference Mazmanian, Round and Kasper2008). Many of our 25 most cited papers adopt this loose definition (e.g., Bercik et al. Reference Bercik, Denou, Collins, Jackson, Lu, Jury, Deng, Blennerhassett, Macri, McCoy, Verdu and Collins2011a; Hsiao et al. Reference Hsiao, McBride, Hsien, Sharon, Hyde, McCue, Codelli, Chow, Reisman, Petrosino, Patterson and Mazmanian2013; Leclercq et al. Reference Leclercq, Matamoros, Cani, Neyrinck, Jamar, Stärkel, Windey, Tremaroli, Bäckhed, Verbeke, de Timary and Delzenne2014), and the term circulates widely in the MGB literature. However, considering the extensive inter-individual variation between each human microbiome, it is very difficult to define what constitutes a “normal” or “healthy” or “balanced” microbiome (Hooks & O'Malley Reference Hooks and O'Malley2017). With such a loose definition, dysbiosis can mean any change in microbiota between two compared groups of patients or animals. Even assumptions that “reduced diversity” is linked to illness outcomes (e.g., Desbonnet et al. Reference Desbonnet, Clarke, Traplin, O'Sullivan, Crispie, Moloney, Cotter, Dinan and Cryan2015) are problematic, because some disease states are associated with increased diversity (Shade Reference Shade2017; Zaneveld et al. Reference Zaneveld, McMinds and Vega Thurber2017).

Worryingly, one of our 25 most cited papers postulated a role for dysbiosis even when no compositional microbiome differences were found pre- and post-intervention in healthy humans (Tillisch et al. Reference Tillisch, Labus, Kilpatrick, Jiang, Stains, Ebrat, Guyonnet, Legrain-Raspaud, Trotin, Naliboff and Mayer2013). Many papers discussing dysbiosis go on to assume that when microbiome changes and illness co-occur, the causal pathway will be from microbiota to the disease state rather than the other way round, or from another common cause (e.g., Bruce-Keller et al. Reference Bruce-Keller, Salbaum, Luo, Blanchard, Taylor, Welsh and Berthoud2015; Crumeyrolle-Arias et al. Reference Crumeyrolle-Arias, Jaglin, Bruneau, Vancassel, Cardona, Daugé, Naudon and Rabot2014; O'Mahony et al. Reference O'Mahony, Marchesi, Scully, Codling, Ceolho, Quigley, Cryan and Dinan2009). However, some MGB papers are now taking more nuanced perspectives on dysbiosis “causality” (e.g., Ohland et al. Reference Ohland, Kish, Bell, Thiesen, Hotte, Pankiv and Madsen2013; Park et al. Reference Park, Collins, Blennerhassett, Ghia, Verdu, Bercik and Collins2013), and the concept is currently receiving considerable critical attention and retheorizing in the broader microbiome literature (e.g., Hooks & O'Malley Reference Hooks and O'Malley2017; Olesen & Alm Reference Olesen and Alm2016; Shanahan & Quigley Reference Shanahan and Quigley2014; Zaneveld et al. Reference Zaneveld, McMinds and Vega Thurber2017).

Lying behind the whole-community causation issue is an even stronger one, of microbiota “determinism.” By this we mean bold claims that are made about human dependency on microbes for many aspects of health (e.g., metabolic, immune, and neuroendocrine systems – see Bercik et al. Reference Bercik, Denou, Collins, Jackson, Lu, Jury, Deng, Blennerhassett, Macri, McCoy, Verdu and Collins2011a; Neufeld et al. Reference Neufeld, Kang, Bienenstock and Foster2011a). These claims include mental health, to the extent that some MGB review papers even suggest our microbiota “control” and “manipulate” our brains (e.g., Stilling et al. Reference Stilling, Dinan and Cryan2016) or “hijack” our central nervous system (e.g., Alcock et al. Reference Alcock, Maley and Aktipis2014). The ability of microbes to determine what we often consider to be central nervous system capacities and states (mood, cognition, emotion, etc.) is a radical one and is probably employed more for provocation than serious consideration. Almost all MGB papers recognize in their small print the lack of a causal account of how microbiota changes are connected to brain and behavioural states. And yet underlying dramatic suggestions that MGB research does away with free will conceptions (e.g., Lepage et al. Reference Lepage, Leclerc, Joossens, Mondot, Blottière, Raes, Ehrlich and Doré2013) is a more reasoned position that microbes are “benevolent” manipulators, and that evolution has made them so. Can evolutionary theory back up such claims?

7.2. Claims about the evolved benefits of microbiota for brain states

There are numerous MGB articles (including some within the 25 most cited sample) that suggest we have a beneficial relationship with many if not all of our microbiota (e.g., Bailey et al. Reference Bailey, Dowd, Galley, Hufnagle, Allen and Lyte2011; Sudo et al. Reference Sudo, Chida, Aiba, Sonoda, Oyama, Yu, Kubo and Koga2004). The reason for this, according to at least some MGB researchers in the broader literature, is supposedly that our long evolutionary association with microorganisms has eradicated conflict (e.g., Stilling et al. Reference Stilling, Dinan and Cryan2016). In other words, natural selection has selected against competitive relationships in the history of human evolution, and we should therefore find the evolved ways in which to maintain the right “balance” with our microbiota (e.g., Wang & Kasper Reference Wang and Kasper2014).

Many such MGB claims begin with the central example of Toxoplasma gondii as a single organism capable of having manipulative effects on animal brains and behaviour (e.g., Mayer et al. Reference Mayer, Knight, Mazmanian, Cryan and Tillisch2014; Sampson & Mazmanian Reference Sampson and Mazmanian2015; Stilling et al. Reference Stilling, Dinan and Cryan2016). Toxoplasma is a single pathogen, and therefore neither benevolent nor a community, but MGB researchers use it to provide an explanatory template for how microbes manipulate. In the classic account of this parasite's effects, Toxoplasma has evolved to infect cats via rodents, and so the former “manipulates” rodent brains in order to make rodents more likely to be consumed by cats (e.g., Berdoy et al. Reference Berdoy, Webster and Macdonald2000). Changed rodent behaviours include attraction to cat urine and odour. However, there are recognized problems in seeing Toxoplasma as evolved by adaptation to change mouse behaviour (Worth et al. Reference Worth, Lymbery and Thompson2013). More generally, “microbial manipulation” of any host is better explained as a by-product of the interactions between competing microorganisms in the gut environment (Johnson & Foster Reference Johnson and Foster2018). In other words, “manipulation” is a considerable overinterpretation of what the microorganisms are doing and how they have their effects.

But what about the generally beneficial nature of microbiota? Some MGB and other microbiome researchers have argued that a long evolutionary association between humans and their microbiota has led to benefits and no conflict (e.g., Stilling et al. Reference Stilling, Dinan and Cryan2016). Evolutionary theory does not support such beliefs. Communities can be stable and perpetuated over evolutionary time with strongly competitive interactions between different microorganismal populations, and between a human host and the whole microbial community (Coyte et al. Reference Coyte, Schluter and Foster2015). Humans are most parsimoniously understood as an environment for microorganisms, and there are mechanisms of human control and selection over inevitable microbial occupants (Schluter & Foster Reference Schluter and Foster2012). There can be negative or positive interactions, as well as neutral ones, and at the moment, microbiome research is unable to separate them out (though efforts are being made to identify key individual microorganisms for specific diseases). But just as for dysbiosis, thinking of whole communities as bringing about specific brain and behavioural (or other physiological) states is very difficult to justify, even (or perhaps especially) within the embrace of evolutionary reasoning.

7.3. Claims about coevolved developmental impact and critical windows

The “coevolved” nature of developmental programmes and microbiota is also argued by the MGB community, both in the 25 papers we examined most closely and more broadly (e.g., Diaz Heijtz et al. Reference Diaz Heijtz, Wang, Anuar, Qian, Björkholm, Samuelsson, Hibberd, Forssberg and Pettersson2011; Stilling et al. Reference Stilling, Bordenstein, Dinan and Cryan2014). Usually, these mentions of “coevolution” do not employ the term in the same way as evolutionary biologists, for whom coevolution means selected reciprocal genetic changes that have been explicitly identified (e.g., Moran & Sloan Reference Moran and Sloan2015). In MGB research, coevolution simply means it appears as if the organisms have some evolutionary history together. Even in this very loose sense, there are problems. For example, the effects of colonizing GF mouse pups (Diaz Heijtz et al. Reference Diaz Heijtz, Wang, Anuar, Qian, Björkholm, Samuelsson, Hibberd, Forssberg and Pettersson2011) and of probiotic treatments on a maternal infection autism mouse model (Hsiao et al. Reference Hsiao, McBride, Hsien, Sharon, Hyde, McCue, Codelli, Chow, Reisman, Petrosino, Patterson and Mazmanian2013) have contributed to interpretations of “coevolution” producing a critical timing point for microbial participation in host gut and brain development. However, interpretations of a critical developmental period for microbiome colonization clash with other findings showing that the microbial colonization of GF adult rodents brings about the same effects as it does for much younger GF animals (Nishino et al. Reference Nishino, Mikami, Takahashi, Tomonaga, Furuse, Hiramoto, Aiba, Koga and Sudo2013). Findings that only male mice are affected developmentally by microbial manipulations are also problematic for general proposals of species-wide neurodevelopmental roles for microbiota (Clarke et al. Reference Clarke, Grenham, Scully, Fitzgerald, Moloney, Shanahan, Dinan and Cryan2013).

There may also be alternative explanations for apparent critical windows of microbiota effects in animal development. The consequences of manipulating gut microbiota on the physiology and behaviour of an organism may be attributable to more traditionally conceived developmental effects. For example, it is has been shown that GF animals have a more permeable blood-brain interface and larger, but less metabolically active enteric neurons during pre- and postnatal development (Braniste et al. Reference Braniste, Al-Asmakh, Kowal, Anuar, Abbaspour, Tóth, Korecka, Bakocevic, Ng, Kundu, Gulyás, Halldin, Hultenby, Nilsson, Hebert, Volpe, Diamond and Pettersson2014; Dupont et al. Reference Dupont, Jervis and Sprinz1965). Given that the enteric nervous system and the blood-brain barrier are essential for the normal functioning of gut and brain, it would not, therefore, be surprising to observe atypical behaviour in an adult animal with abnormal development of these systems. However, any behavioural changes do not imply that gut microbiota “control” or “drive” a particular behaviour, but merely that the presence of microbes in the gut may constitute environmental signals to which the developing animal responds by putting in place an enteric neuronal network and a blood-brain barrier.

The adoption of evolutionary-developmental (evo-devo) frameworks in MGB research has also led to studies intimating that if microbes have a big effect on brain development, this must also be occurring prenatally. Some MGB researchers hint that there are large numbers of microorganisms in utero, and that these organisms are having a pre-birth impact on the foetal brain (e.g., Borre et al. Reference Borre, O'Keeffe, Clarke, Stanton, Dinan and Cryan2014; O'Mahony et al. Reference O'Mahony, Clarke, Dinan and Cryan2017). Yet if they are, current orthodoxy of a mostly sterile pre-birth state would have to be revised.

Recent analysis casts considerable doubt on the potential for in utero colonization and concludes that apparent findings of such colonization are artefactual (Perez-Muñoz et al. Reference Perez-Muñoz, Arrieta, Ramer-Tait and Walter2017). Low-microbial biomass samples, such as those extracted from placenta, yield a similar composition to those from negative controls and are, in fact, dependent on the type or even batch of the kit used to extract and examine the DNA sample. This is the so-called kit-ome problem (see Kim et al. Reference Kim, Hofstaedter, Zhao, Mattei, Tanes, Clarke, Lauder, Sherrill-Mix, Chehoud, Kelsen, Conrad, Collman, Baldassano, Bushman and Bittinger2017a). Artefacts such as these can be more straightforward explanations of what are otherwise very surprising microbiome findings. That said, we have no doubt that something is going on in an evo-devo sense with microbiota and brains. But expecting simple and straightforward findings and linear causal accounts of these interactions does not seem to us realistic, given existing knowledge and methodological sophistication in standard developmental research. There are other oversimplified causal stories that MGB research needs to confront, and chief amongst them are claims about probiotics.

7.4. Probiotic issues

Using the template of the Sudo et al. (Reference Sudo, Chida, Aiba, Sonoda, Oyama, Yu, Kubo and Koga2004) study, many subsequent MGB projects (including those in the 25 most cited papers) have made interventions with probiotics on mice and humans and claimed that probiotic interactions with indigenous microbiota affect physiology and behaviour (e.g., Diaz Heijtz et al. Reference Diaz Heijtz, Wang, Anuar, Qian, Björkholm, Samuelsson, Hibberd, Forssberg and Pettersson2011; Lyte Reference Lyte2011; Messaoudi et al. Reference Messaoudi, Lalonde, Violle, Javelot, Desor, Nejdi, Bisson, Rougeot, Pichelin, Cazaubiel and Cazaubiel2011; Slykerman et al. Reference Slykerman, Hood, Wickens, Thompson, Barthow, Murphy, Kang, Rowden, Stone, Crane, Stanley, Abels, Purdie, Maude and Mitchell2017; Steenbergen et al. Reference Steenbergen, Sellaro, van Hemert, Bosch and Colzato2015; see Table 1). Often this interaction is conceptualized as the abnormal or “dysbiotic” microbiota being “normalized” by the probiotic (e.g., Ait-Belgnaoui et al. Reference Ait-Belgnaoui, Durand, Cartier, Chaumaz, Eutamene, Ferrier, Houdeau, Fioramonti, Bueno and Theodorou2012). However, probiotics are a much contested form of intervention. Meta-analyses are equivocal at best about probiotics having positive effects on healthy humans, and their impact is documented for only a few specific disease states (Huang et al. Reference Huang, Wang and Hu2016; McKean et al. Reference McKean, Naug, Nikbakht, Amiet and Colson2017). At least two randomized controlled trials have found no human effects from probiotic bacteria on human mood or mental health (Kelly et al. Reference Kelly, Allen, Temko, Hutch, Kennedy, Farid, Murphy, Boylan, Bienenstock, Cryan, Clarke and Dinan2017; Romijn et al. Reference Romijn, Rucklidge, Kuijer and Frampton2017), whereas a recent meta-analysis (Ng et al. Reference Ng, Peters, Ho, Lim and Yeo2017) observed no general mood improvement after using probiotics and only a small effect in patients with mild to moderate depressive symptoms. Concerns have also been raised about the potentially negative alteration of microbiota by probiotics (Slashinski et al. Reference Slashinski, McCurdy, Achenbaum, Whitney and McGuire2012). However, mouse studies do seem to show probiotics having consistent effects on behaviour (Wang et al. Reference Wang, Lee, Braun and Enck2016), and such findings continue to galvanize the MGB field.

Even if probiotics do have positive effects on guts and brains, some studies show this may not be happening through alterations of the microbiome composition (Kristensen et al. Reference Kristensen, Bryrup, Allin, Nielsen, Hansen and Pedersen2016; McNulty et al. Reference McNulty, Yatsunenko, Hsiao, Faith, Muegge, Goodman, Henrissat, Oozeer, Cools-Portier, Gobert, Chervaux, Knights, Lozupone, Knight, Duncan, Bain, Muehlbauer, Newgard, Heath and Gordon2011). Recent work shows that probiotics do not reliably colonize mouse guts, and do so only to a limited extent for humans (Zmora et al. Reference Zmora, Zilberman-Schapira, Suez, Mor, Dori-Bachash, Bashiardes, Kotler, Zur, Regev-Lehavi, Brik, Federici, Cohen, Linevsky, Rothschild, Moor, Ben-Moshe, Harmelin, Itzkovitz, Maharshak, Shibolet, Shapiro, Pevsner-Fischer, Sharon, Halpern, Segal and Elinav2018). Sampson and Mazmanian (Reference Sampson and Mazmanian2015) account for such complications by suggesting more indirect causal routes: “behavioral and neurological changes may not necessarily be a direct function of the specific species of bacteria within the probiotic treatment; rather, microbial-mediated effects on emotion may be due to broader functionality of the community of symbiotic bacteria in the gut” (p. 568). Claims like these fall into what we call the whole-system causation problem that is central to the “dysbiosis” problem (see sect. 7.1). They are very difficult claims to test, especially in a medical context. One of the most cited MGB papers, Ohland et al. (Reference Ohland, Kish, Bell, Thiesen, Hotte, Pankiv and Madsen2013, p. 1746), carefully concludes:

It is clear that diet and probiotics interact at several different levels to alter host physiology. It is likely that not only do the existing gut microbes of the host alter functionality of any given probiotic, but also the diet of the host can influence probiotic effects through both direct and indirect mechanisms. These differences in probiotic effects due to diet and genotype demonstrate that it is essential to investigate probiotics in a complex model to fully understand how they modulate host physiology in order to properly apply them to improve human health.

Regardless of how sketchy the current causal picture is of microbiota and mental health, probiotics are a commercial goldmine. They are the basis of an industry that already (in 2015) earns 35 billion dollars per year (Jabr Reference Jabr2017). To gain a closer view of the appeal of probiotics, we examined patenting trends for microbiota and probiotics. A very high proportion of microbiota/microbiome patents are for probiotics (see sect. 7 in the Supplementary Material). Commercial investment in probiotics is increasing (Jabr Reference Jabr2017; Olle Reference Olle2013), as is academic patenting activity related to probiotic and other microbiota-based therapies (Supplementary Fig. S1). Nestlé, the biggest food company in the world, leads the way with probiotic patents and patent applications in the European Patent Office; Danone, another large food company with many dairy-based products, ranks fourth (Supplementary Fig. S2).

With its simple cause-effect hints (“take probiotics and cure yourself”), MGB research is likely to attract even more commercial attention and funding. Perhaps maintaining this appeal is part of the reason so many MGB studies repeat the basic recipe of probiotic-based intervention as the single “microbiome” method. In this research environment, single-study findings of no effect from probiotics are simply less likely to be published (although meta-analyses and systematic reviews with negative findings do find publishing forums), and the complex models urged by some researchers will have limited appeal. However, as the occasional commentator has noted (e.g., Olle Reference Olle2013), focusing on a few classic probiotic strains – identified more than a century ago by much cruder methods – seems an unduly narrow focus given how microbiome research is normally about highlighting community-wide microbial diversity and interaction. But perhaps for this very reason probiotics remain popular. They enable straightforward experimentation, by appearing to cut through complex interactions and thus suggest that simple, non-harmful treatments are possible, even for conditions as resistant to conventional interventions as autism (de Theije et al. Reference de Theije, Wopereis, Ramadan, van Eijndthoven, Lambert, Knol, Garssen, Kraneveld and Oozeer2014). This simplicity is important for the public uptake of MGB and other microbiome research.

7.5. Science communication issues

Human microbiome research has captured the public imagination. It is a very popular professional media topic. A simple search for “gut microbiota” in the Factiva press database retrieves almost 1,500 publications. Even when narrowed down to a “microbiota gut brain” focus, the search still yields more than 300 press publications (see sect. 8 in the Supplementary Material, especially Fig. S3, for details). Less than a third of these press articles contain elements of caution or scepticism, and most are accompanied by very enthusiastic and optimistic claims. Generally, these articles make simple and encouraging reports on microbiome research and its potential impact on physical and mental health (e.g., “Pathogens in the stomach alter the brain's development and may increase an individual's risk of suffering from [autism] spectrum disorder” [Thompson Reference Thompson2017]). A common template is to highlight dietary change (including probiotics) as a “natural” means of changing the microbiome, and thus host health status (e.g., “Taking probiotics and adopting a gluten-free lifestyle may improve [autism] sufferers' social behaviour and ability to express emotions” [Thompson Reference Thompson2017]).

A valuable lesson for press releases about research can be learned from early microbiome research on obesity (e.g., Turnbaugh et al. Reference Turnbaugh, Ley, Mahowald, Magrini, Mardis and Gordon2006). Numerous studies, both experimental and bioinformatic, found associations between certain proportions of microorganismal groups and obesity. However, as these studies accumulated, this allowed meta-analyses and systematic reviews to be conducted, and these earlier findings fell away (Duvallet et al. Reference Duvallet, Gibbons, Gurry, Irizarry and Alm2017; Sze & Schloss Reference Sze and Schloss2016). Initial findings, although widespread, were from small samples, with hidden variations in background conditions (Schloss Reference Schloss2018). As we already noted, high inter-individual variability means large samples are required to make meaningful findings. Apparent effects in the obesity case turned out not to be real. Such developments in a new field are not surprising. It takes an accumulation of studies to allow meta-analyses to be conducted, and once they are, the field can correct itself.

However, even if a field manages to correct itself, systematic analyses of press articles have shown that public media material, including that produced by academic public relations offices, often focuses on initial spectacular findings. These early findings are often obtained from relatively small samples and are promissory rather than enduring (Gonon et al. Reference Gonon, Bezard and Boraud2011; Reference Gonon, Konsman, Cohen and Boraud2012). Although early dramatic findings and press coverage can help attract funds to fledgling fields, and rapidly inform the public about potential avenues of treatment, the downsides are misinformation, unrealistic expectations, and eventual public and political backlash. The last is especially likely if initial findings cannot be translated into accessible therapies quite as readily as press releases might suggest (Hanage Reference Hanage2014).

But professional media are probably of less magnitude in this potentially misleading communication than is the large number of social media posts discussing microbiomes and health generally, and mental health in particular. Although we did not systematically survey blogs, tweets, and other such media, we did examine the first 50 Google hits for searches using gut + brain + microbiome (see Supplementary Table 8). Additionally, we performed a survey of Twitter posts of news articles in 2017 (see Supplementary Table 9). Although many of these online materials refer to actual research, they rarely do so critically. At most, they acknowledge that much more research has to be done. Notable exceptions within our small sample are an opinion piece cautioning against blanket belief in the efficacy of probiotics (DiSalvo Reference DiSalvo2017) and a book review raising questions about the simplicity of the “psychobiotic” approach (Fleming Reference Fleming2017; many reader comments are sceptical too).

The majority of the posts and shared news articles we surveyed suggest that new microbiome-related mental health treatments are just around the corner. Some websites and Twitter accounts promote probiotic and other dietary interventions as replacements for conventional psychiatric treatments. Many of these alternative “treatments” accord with standard nutritional and lifestyle guidelines (eat more fresh and less processed food, less fat and sugar, and more fibre; get more exercise; avoid stress). These are reasonable and no doubt helpful recommendations, regardless of how idiosyncratically some of them may be phrased on Twitter. What is concerning, however, is how this very ordinary dietary advice can be proposed as the solution to many mental health conditions. Even though clear cause-effect links between diet-altered microbiota composition and bodily or mental status are unknown, these gaps themselves leave room for the sentiment that it is all just “common sense” and that science is finally catching up to what everyone already knew in his or her gut anyway. Some MGB papers in the broader literature appear to endorse this way of thinking (e.g., Cowan et al. Reference Cowan, Hoban, Ventura-Silva, Dinan, Clarke and Cryan2018) and may even sign up for other dubious health claims floating about in the public sphere. For example, using “leaky gut” language when it is not medically recognized as the basis of any disorder, let alone as a major causative agent of autism syndromes (Quigley Reference Quigley2016; Rao & Gershon Reference Rao and Gershon2016), is harnessing science to the fortune of what may be a medical fad.

As Perez-Muñoz et al. (Reference Perez-Muñoz, Arrieta, Ramer-Tait and Walter2017) argue, when they debunk claims of in utero or placental colonization:

Today, scientific findings can move freely from professional journals into the public realm (e.g., through social media), often before the scientific community has thoroughly discussed and vetted the evidence … it is our responsibility [as scientists] to debate these controversial topics and facilitate the self-correction process. Failure to do so may ultimately compromise human health, damage scientific creditability [sic], and potentially contribute to the erosion of the public's trust in science. (p. 15)

We suggest that human microbiome research in general (Hanage Reference Hanage2014) and MGB research specifically are at a point where careful reflection on the broader reception of the science would be highly appropriate.

8. Summarizing our findings

To its credit, MGB research is driven by hypothesis testing, but it mostly proposes and confirms loose conjectures about microbial involvement in brain and behavioural states. Microbiome research (outside MGB) is very technology driven and often fishes around after analysis for some sort of hypothesis that might reasonably be based on the data. Neither extreme of this continuum of practice is desirable for the maturation of microbiome research. In fact, we could see in MGB research the potential to integrate and balance these two ways of doing science. Very importantly, this merger would bring more microbiome depth to MGB research, which our analysis shows is missing and misunderstood.

We also showed how MGB research has many other compounding methodological and interpretive issues. But might all the issues we have identified just be signs of a young field? Will it not get better all of its own accord, given enough time? We agree it is important not to inhibit new approaches as they develop. But a strong foundation seems important for future development, rather than ongoing reproduction of a rough-and-ready approach. We have taken a critical approach to this emerging field, partly because we see the same claims repeated over and over again. They achieve a wider reach with every iteration. Using evolutionary, ecological, microbiological, neurological, immunological, biochemical, genetic, molecular, and developmental perspectives to bolster a narrow band of results both overreaches and also displays limited acquaintance with some of the well-established knowledge in these fields. These limitations matter not only for the future of a field, but also for the status of scientific activity in these challenging times. As we suggest and others have argued (e.g., Hanage Reference Hanage2014; Perez-Muñoz et al. Reference Perez-Muñoz, Arrieta, Ramer-Tait and Walter2017), overblown claims damage the credibility of the field and cause harm to the general social reception of science.

A topic worthy of further social scientific investigation is why microbiome research in general is so popular with the public, and whether public perspectives on microbiome research are changing how people think about health, including mental health. We speculate that reasons for the public uptake of microbiome research findings, including MGB, are to do with its perceived “naturalness” and the “holism” of the science, as well as the strong potential for microbiome-related therapies to be self-administered and even “DIY” rather than imposed by technical experts. There are many good aspects to any such trends. But MGB research should be aware of these tendencies and their possible relationships with anti-scientific claims (e.g., anti-vaccination; anti-psychotropic medication). It could be well worth working with relevant public health and media experts on how to communicate this exciting body of work responsibly.

9. Conclusions and future directions

Despite the critical picture we have painted, we see MGB research as a field full of promise, with important implications for understanding the relationship between the brain and the rest of the body. Existing MGB findings point to an ongoing need for more connected research that is able to investigate the complex interactions occurring in multipathway systems. Expecting cure-alls to emerge from these early days in which the puzzle pieces have barely been recognized, let alone joined up, seems contradictory to the spirit we assume to be motivating MGB inquiry. Our findings indicate the tension between a field-wide recognition of complex networks of causes and effects versus expectations of a simple all-efficacious treatment. As we noted in the introduction, our critical overview of MGB research is from outside the field itself and does not presume it can provide the detailed advice necessary to lead the field forward. This has to come from within the field. Nevertheless, we can use our findings of the current state of the field to propose some general pointers about how the field might develop and what it should avoid in that development.

9.1. What is known?

Perhaps the clearest general finding from MGB and the encompassing field of microbiome research is that microbiota are implicated in a wide range of ecosystem activities, some of which take place in human and other animal bodies and may be of considerable importance for understanding health and disease. Some of these connections are surprising, even if foreshadowed by earlier research (see sect. 2), and, if worked out experimentally and in clinical trials, could transform treatment options for ill humans. There do indeed seem to be links between microbes and mental health states, but they are extensively mediated by developmental, immunological, and metabolic processes that are in turn affected by environmental factors. Quite what these microbiome connections entail is the central question, and revealing the nature of any causal processes involving microbiota is what all MGB and other microbiome studies ultimately aim to do. Many researchers in MGB are now trying to fill the causal gaps and narrow down how microbiota or probiotics change mental health.

9.2. What is improving?

Several MGB and other microbiome papers in recent years have urged more rigorous experimental design, with appropriate positive and negative controls and adequate statistical power to allow the identification of cause and effect relationships and point to mechanistic explanations (e.g., Bruce-Keller et al. Reference Bruce-Keller, Salbaum and Berthoud2018; Lyte Reference Lyte2011; Schloss Reference Schloss2018). More sophisticated microbiota sampling and analysis will help us understand which groups of organisms are contributing to putative effects (Knight et al. Reference Knight, Vrbanac, Taylor, Aksenov, Callewaert, Debelius, Gonzalez, Kosciolek, McCall, McDonald, Melnik, Morton, Navas, Quinn, Sanders, Swafford, Thompson, Tripathi, Xu, Zaneveld, Zhu, Caporaso and Dorrestein2018). Models that capture such interactions and their dynamics over time are going to be crucial, and some are already developed for broader microbiome research (e.g., Bucci & Xavier Reference Bucci and Xavier2014).

Integrating multiple levels of causal influence in producing any kind of disease is always challenging, but if there is one thing microbiome research brings to the fore, it is awareness of the challenges in making causal claims about complex systems. The earlier rush to identify promising causal relationships in MGB research, and simplistically attribute large-scale effects to “the microbiome,” or one-off probiotic interventions, can most constructively be understood as heuristic strategies that await more rigorous inquiry. There is now sufficient background knowledge to allow the refinement of hypotheses about microbiota relationships, and placeholder claims about causality can be put to the test.

9.3. What should be stopped?

Although we see many positive developments along methodological lines in MGB research, it is still accompanied by large helpings of overinterpretation, even if these come with a sprinkling of caution. Sometimes, it seems as if cautionary statements are used as liability limitation clauses in the ongoing promotion of the research (this is what we labelled in sect. 7 as “double-dipping”). Helpful as reviews may be to introduce non-experts to an emerging field, the wholesale marketing of MGB research in such a prolific review literature may “oversell” currently limited findings. Being more strategic about how the field is promoted, within and without science, could have long-run dividends that MGB researchers may want to consider.

Supplementary

To view supplementary material for this article, please visit https://doi.org/10.1017/S0140525X18002133.

Acknowledgements

KBH and MAO acknowledge support from the French government via the “Investments for the Future” Programme, IdEx Bordeaux (ANR-10-IDEX-03-02). JPK was financially supported in 2018 by the CNRS as part of its “Osez l'interdisciplinarité!” programme.

References

Ait-Belgnaoui, A., Colom, A., Braniste, V., Ramalho, L., Marrot, A., Cartier, C., Houdeau, E., Theodorou, V. & Tompkins, T. (2014) Probiotic gut effect prevents the chronic psychological stress-induced brain activity abnormality in mice. Neurogastroenterology and Motility 26(4):510–20. https://doi.org/10.1111/nmo.12295.Google Scholar
Ait-Belgnaoui, A., Durand, H., Cartier, C., Chaumaz, G., Eutamene, H., Ferrier, L., Houdeau, E., Fioramonti, J., Bueno, L. & Theodorou, V. (2012) Prevention of gut leakiness by a probiotic treatment leads to attenuated HPA response to an acute psychological stress in rats. Psychoneuroendocrinology 37(11):1885–95. Available at: https://doi.org/10.1016/j.psyneuen.2012.03.024.Google Scholar
Alcock, J., Maley, C. C. & Aktipis, C. A. (2014) Is eating behavior manipulated by the gastrointestinal microbiota? Evolutionary pressures and potential mechanisms. BioEssays 36:940–49. Available at: https://doi.org/10.1002/bies.201400071.Google Scholar
Arrieta, M.-C., Walter, J. & Finlay, B. B. (2016) Human microbiota-associated mice: A model with challenges. Cell Host & Microbe 19:575–78. Available at: https://doi.org/10.1016/j.chom.2016.04.014.Google Scholar
Asano, Y., Hiramoto, T., Nishino, R., Aiba, Y., Kimura, T., Yoshihara, K., Koga, Y. & Sudo, N. (2012) Critical role of gut microbiota in the production of biologically active, free catecholamines in the gut lumen of mice. AJP Gastrointestinal and Liver Physiology 303:G128895. Available at: https://doi.org/10.1152/ajpgi.00341.2012.Google Scholar
Aziz, Q. & Thompson, D. G. (1998) Brain-gut axis in health and disease. Gastroenterology 114:559–78. Available at: https://doi.org/10.1016/S0016-5085(98)70540-2.Google Scholar
Bailey, M. T., Dowd, S. E., Galley, J. D., Hufnagle, A. R., Allen, R. G. & Lyte, M. (2011) Exposure to a social stressor alters the structure of the intestinal microbiota: Implications for stressor-induced immunomodulation. Brain, Behavior, and Immunity 25(3):397407. Available at: https://doi.org/10.1016/j.bbi.2010.10.023.Google Scholar
Bajaj, J. S., Heuman, D. M., Sanyal, A. J., Hylemon, P. B., Sterling, R. K., Stravitz, R. T., Fuchs, M., Ridlon, J. M., Daita, K., Monteith, P., Noble, N. A., White, M. B., Fisher, A., Sikaroodi, M., Rangwala, H. & Gillevet, P. M. (2013) Modulation of the metabiome by rifaximin in patients with cirrhosis and minimal hepatic encephalopathy. PLoS ONE 8:e60042. Available at: https://doi.org/10.1371/journal.pone.0060042.Google Scholar
Bassi, G. S., Kanashiro, A., Santin, F. M., de Souza, G. E. P., Nobre, M. J. & Coimbra, N. C. (2012) Lipopolysaccharide-induced sickness behaviour evaluated in different models of anxiety and innate fear in rats. Basic & Clinical Pharmacology & Toxicology 110:359–69. Available at: https://doi.org/10.1111/j.1742-7843.2011.00824.x.Google Scholar
Belzung, C. & Griebel, G. (2001) Measuring normal and pathological anxiety-like behaviour in mice: A review. Behavioural Brain Research 125:141–49. Available at: https://doi.org/10.1016/S0166-4328(01)00291-1.Google Scholar
Bercik, P., Denou, E., Collins, J., Jackson, W., Lu, J., Jury, J., Deng, Y., Blennerhassett, P., Macri, J., McCoy, K. D., Verdu, E. F. & Collins, S. M. (2011a) The intestinal microbiota affect central levels of brain-derived neurotropic factor and behavior in mice. Gastroenterology 141:599609, 609.e1–3. Available at: https://doi.org/10.1053/j.gastro.2011.04.052.Google Scholar
Berdoy, M., Webster, J. P. & Macdonald, D. W. (2000) Fatal attraction in rats infected with Toxoplasma gondii. Proceedings of the Royal Society B: Biological Sciences 267:1591–94. Available at: https://doi.org/10.1098/rspb.2000.1182.Google Scholar
Bik, E. M. (2016) The hoops, hopes, and hypes of human microbiome research. Yale Journal of Biology and Medicine 89:363–73. Available at: http://www.ncbi.nlm.nih.gov/pubmed/27698620.Google Scholar
Bluthé, R. M., Dantzer, R. & Kelley, K. W. (1992) Effects of interleukin-1 receptor antagonist on the behavioral effects of lipopolysaccharide in rat. Brain Research 573:318–20. Available at: http://www.ncbi.nlm.nih.gov/pubmed/1387028.Google Scholar
Borre, Y. E., O'Keeffe, G. W., Clarke, G., Stanton, C., Dinan, T. G. & Cryan, J. F. (2014) Microbiota and neurodevelopmental windows: Implications for brain disorders. Trends in Molecular Medicine 20(9):509–18. Available at: https://doi.org/10.1016/j.molmed.2014.05.002.Google Scholar
Bourin, M., Petit-Demoulière, B., Dhonnchadha, B. N. & Hascöet, M. (2007) Animal models of anxiety in mice. Fundamental & Clinical Pharmacology 21:567–74. Available at: https://doi.org/10.1111/j.1472-8206.2007.00526.x.Google Scholar
Braniste, V., Al-Asmakh, M., Kowal, C., Anuar, F., Abbaspour, A., Tóth, M., Korecka, A., Bakocevic, N., Ng, L. G., Kundu, P., Gulyás, B., Halldin, C., Hultenby, K., Nilsson, H., Hebert, H., Volpe, B. T., Diamond, B. & Pettersson, S. (2014) The gut microbiota influences blood-brain barrier permeability in mice. Science Translational Medicine 6:263ra158. Available at: https://doi.org/10.1126/scitranslmed.3009759.Google Scholar
Bravo, J. A., Forsythe, P., Chew, M. V., Escaravage, E., Savignac, H. M., Dinan, T. G., Bienenstock, J. & Cryan, J. F. (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 USA 108(38):16050–55. Available at: https://doi.org/10.1073/pnas.1102999108.Google Scholar
Brown, R., Price, R. J., King, M. G. & Husband, A. J. (1990) Are antibiotic effects on sleep behavior in the rat due to modulation of gut bacteria? Physiology & Behavior 48:561–65.Google Scholar
Bruce-Keller, A. J., Salbaum, J. M. & Berthoud, H.-R. (2018) Harnessing gut microbes for mental health: Getting from here to there. Biological Psychiatry 83:214–23. Available at: https://doi.org/10.1016/j.biopsych.2017.08.014.Google Scholar
Bruce-Keller, A. J., Salbaum, J. M., Luo, M., Blanchard, E., Taylor, C. M., Welsh, D. A. & Berthoud, H.-R. (2015) Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity. Biological Psychiatry 77:607–15. Available at: https://doi.org/10.1016/j.biopsych.2014.07.012.Google Scholar
Bucci, V. & Xavier, J. B. (2014) Towards predictive models of the human gut microbiome. Journal of Molecular Biology 426:3907–16. Available at: https://doi.org/10.1016/j.jmb.2014.03.017.Google Scholar
Button, K. S., Ioannidis, J. P. A., Mokrysz, C., Nosek, B. A., Flint, J., Robinson, E. S. J. & Munafò, M. R. (2013) Power failure: Why small sample size undermines the reliability of neuroscience. Nature Reviews Neuroscience 14:365–76. Available at: https://doi.org/10.1038/nrn3475.Google Scholar
Cannon, W. B. (1909) The influence of emotional states on the functions of the alimentary canal. American Journal of the Medical Sciences 137:480–87.Google Scholar
Cannon, W. B. (1911) The mechanical factors of digestion. Edward Arnold.Google Scholar
Chedid, L. & Boyer, F. (1953) [Action of adrenaline on the effects of a bacterial endotoxin]. Comptes Rendus Des Seances de La Societe de Biologie et de Ses Filiales 147:1742–44. Available at: http://www.ncbi.nlm.nih.gov/pubmed/13150667.Google Scholar
Christian, L. M., Galley, J. D., Hade, E. M., Schoppe-Sullivan, S., Kamp Dush, C. & Bailey, M. T. (2015) Gut microbiome composition is associated with temperament during early childhood. Brain, Behavior, and Immunity 45:118–27. Availability at: https://doi.org/10.1016/j.bbi.2014.10.018.Google Scholar
Clarke, G., Grenham, S., Scully, P., Fitzgerald, P., Moloney, R. D., Shanahan, F., Dinan, T. G. & Cryan, J. F. (2013) The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Molecular Psychiatry 18(6):666–73. Available at: https://doi.org/10.1038/mp.2012.77.Google Scholar
Colquhoun, D. (2014) An investigation of the false discovery rate and the misinterpretation of p-values. Royal Society Open Science 1:140216. Available at: https://doi.org/10.1098/rsos.140216.Google Scholar
Commons, K. G., Cholanians, A. B., Babb, J. A. & Ehlinger, D. G. (2017) The rodent forced swim test measures stress-coping strategy, not depression-like behavior. ACS Chemical Neuroscience 8:955960. Available at: https://doi.org/10.1021/acschemneuro.7b00042.Google Scholar
Cowan, C. S. M., Hoban, A. E., Ventura-Silva, A. P., Dinan, T. G., Clarke, G. & Cryan, J. F. (2018) Gutsy moves: The amygdala as a critical node in microbiota to brain signaling. BioEssays 40:1700172. Available at: https://doi.org/10.1002/bies.201700172.Google Scholar
Coyte, K. Z., Schluter, J. & Foster, K. R. (2015) The ecology of the microbiome: Networks, competition, and stability. Science 350:663–66. Available at: https://doi.org/10.1126/science.aad2602.Google Scholar
Crumeyrolle-Arias, M., Jaglin, M., Bruneau, A., Vancassel, S., Cardona, A., Daugé, V., Naudon, L. & Rabot, S. (2014) Absence of the gut microbiota enhances anxiety-like behavior and neuroendocrine response to acute stress in rats. Psychoneuroendocrinology 42:207–17. Available at: https://doi.org/10.1016/j.psyneuen.2014.01.014.Google Scholar
Desbonnet, L., Clarke, G., Shanahan, F., Dinan, T. G. & Cryan, J. F. (2014) Microbiota is essential for social development in the mouse. Molecular Psychiatry 19:146–48. Available at: https://doi.org/10.1038/mp.2013.65.Google Scholar
Desbonnet, L., Clarke, G., Traplin, A., O'Sullivan, O., Crispie, F., Moloney, R. D., Cotter, P. D., Dinan, T. G. & Cryan, J. F. (2015) Gut microbiota depletion from early adolescence in mice: Implications for brain and behaviour. Brain, Behavior, and Immunity 48:165–73. Available at: https://doi.org/10.1016/j.bbi.2015.04.004.Google Scholar
de Theije, C. G., Wopereis, H., Ramadan, M., van Eijndthoven, T., Lambert, J., Knol, J., Garssen, J., Kraneveld, A. D. & Oozeer, R. (2014) Altered gut microbiota and activity in a murine model of autism spectrum disorders. Brain, Behavior, and Immunity 37:197206. Available at: https://doi.org/10.1016/j.bbi.2013.12.005.Google Scholar
de Vos, W. M. & de Vos, E. A. J. (2012) Role of the intestinal microbiome in health and disease: From correlation to causation. Nutrition Reviews 70(S1):S4556. Available at: https://doi.org/10.1111/j.1753-4887.2012.00505.x.Google Scholar
Diaz Heijtz, R., Wang, S., Anuar, F., Qian, Y., Björkholm, B., Samuelsson, A., Hibberd, M. L., Forssberg, H. & Pettersson, S. (2011) Normal gut microbiota modulates brain development and behavior. Proceedings of the National Academy of Sciences USA 108:3047–52. Available at: https://doi.org/10.1073/pnas.1010529108.Google Scholar
Dinan, T. G., Stanton, C. & Cryan, J. F. (2013) Psychobiotics: A novel class of psychotropic. Biological Psychiatry 74:720–26. Available at: https://doi.org/10.1016/j.biopsych.2013.05.001.Google Scholar
DiSalvo, D. (2017) Science is showing how gut bacteria affect the brain, but don't bother taking probiotics yet. Forbes, August 27. Available at: https://www.forbes.com/sites/daviddisalvo/2017/08/27/science-is-showing-how-gut-bacteria-affect-the-brain-but-dont-bother-taking-probiotics-yet.Google Scholar
Dupont, J. R., Jervis, H. R. & Sprinz, H. (1965) Auerbach's plexus of the rat cecum in relation to the germfree state. Journal of Comparative Neurology 125:1118. Available at: http://www.ncbi.nlm.nih.gov/pubmed/5866590.Google Scholar
Duvallet, C., Gibbons, S. M., Gurry, T., Irizarry, R. A. & Alm, E. J. (2017) Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nature Communications 8:1784. Available at: https://doi.org/10.1038/s41467-017-01973-8.Google Scholar
Eckburg, P. B., Bik, E. M., Bernstein, C. N., Purdom, E., Dethlefsen, L., Sargent, M., Gill, S. R., Nelson, K. E. & Relman, D. A. (2005) Diversity of the human intestinal microbial flora. Science 308:1635–38. Available at: https://doi.org/10.1126/science.1110591.Google Scholar
Eik-Nes, K. B. & Samuels, L. T. (1958) Metabolism of cortisol in normal and stressed dogs. Endocrinology 63:8288. Available at: https://doi.org/10.1210/endo-63-1-82.Google Scholar
Eisen, J. A. (2017) Microbiomania and “overselling the microbiome.” The Tree of Life (blog). Available at: https://phylogenomics.blogspot.com/p/blog-page.html.Google Scholar
Eisenstein, M. (2016) Microbiome: Bacterial broadband. Nature 533:S104106. Available at: https://doi.org/10.1038/533S104a.Google Scholar
Ennaceur, A. (2014) Tests of unconditioned anxiety – Pitfalls and disappointments. Physiology & Behavior 135:5571. Available at: https://doi.org/10.1016/j.physbeh.2014.05.032.Google Scholar
Evans, D. G., Miles, A. A. & Niven, J. S. F. (1948) The enhancement of bacterial infections by adrenaline. British Journal of Experimental Pathology 29:2039. Available at: http://www.ncbi.nlm.nih.gov/pubmed/18865102.Google Scholar
Falony, G., Joossens, M., Vieira-Silva, S., Wang, J., Darzi, Y., Faust, K., Kurilshikov, A., Bonder, M. J., Valles-Colomer, M., Vandeputte, D., Tito, R. Y., Chaffron, S., Rymenans, L., Verspecht, C., De Sutter, L., Lima-Mendez, G., D'Hoe, K., Jonckheere, K., Homola, D., Garcia, R., Tigchelaar, E. F., Eeckhaudt, L., Fu, J., Henckaerts, L., Zhernakova, A., Wijmenga, C. & Raes, J. (2016) Population-level analysis of gut microbiome variation. Science 352(6285):560–64. Available at: https://doi.org/10.1126/science.aad3503.Google Scholar
Fleming, A. (2017) Is your gut microbiome the key to health and happiness? Guardian, November 6. Available at: https://www.theguardian.com/lifeandstyle/2017/nov/06/microbiome-gut-health-digestive-system-genes-happiness.Google Scholar
Forsythe, P., Kunze, W. & Bienenstock, J. (2016) Moody microbes or fecal phrenology: What do we know about the microbiota-gut-brain axis? BMC Medicine 14:58. Available at: https://doi.org/10.1186/s12916-016-0604-8.Google Scholar
Foster, J. A. & McVey Neufeld, K.-A. (2013) Gut-brain axis: How the microbiome influences anxiety and depression. Trends in Neurosciences 36(5):305–12. Available at: https://doi.org/10.1016/j.tins.2013.01.005.Google Scholar
Gareau, M. G., Wine, E., Rodrigues, D. M., Cho, J. H., Whary, M. T., Philpott, D. J., Macqueen, G. & Sherman, P. M. (2011) Bacterial infection causes stress-induced memory dysfunction in mice. Gut 60:307–17. Available at: https://doi.org/10.1136/gut.2009.202515.Google Scholar
Gastroenterology (1980) Notices: International symposium on the brain-gut axis. Gastroenterology 79(4):783.Google Scholar
Gevers, D., Kugathasan, S., Denson, L. A., Vázquez-Baeza, Y., Van Treuren, W., Ren, B., Schwager, E., Knights, D., Song, S. J., Yassour, M., Morgan, X. C., Kostic, A. D., Luo, C., González, A., McDonald, D., Haberman, Y., Walters, T., Baker, S., Rosh, J., Stephens, M., Heyman, M., Markowitz, J., Baldassano, R., Griffiths, A., Sylvester, F., Mack, D., Kim, S., Crandall, W., Hyams, J., Huttenhower, C., Knight, R. & Xavier, R. J. (2014) The treatment-naive microbiome in new-onset Crohn's disease. Cell Host & Microbe 15:382–92. Available at: https://doi.org/10.1016/j.chom.2014.02.005.Google Scholar
Goehler, L. E., Park, S. M., Opitz, N., Lyte, M. & Gaykema, R. P. A. (2008) Campylobacter jejuni infection increases anxiety-like behavior in the holeboard: Possible anatomical substrates for viscerosensory modulation of exploratory behavior. Brain, Behavior, and Immunity 22:354–66. Available at: https://doi.org/10.1016/j.bbi.2007.08.009.Google Scholar
Gold, N. I., Singleton, E., Macfarlane, D. A. & Moore, F. D. (1958) Quantitative determination of the urinary cortisol metabolites, “tetrahydro F,” “allo-tetrahydro F” and “tetrahydro E”: Effects of adrenocorticotropin and complex trauma in the human. Journal of Clinical Investigation 37:813–23. Available at: https://doi.org/10.1172/JCI103669.Google Scholar
Gonon, F., Bezard, E. & Boraud, T. (2011) Misrepresentation of neuroscience data might give rise to misleading conclusions in the media: The case of attention deficit hyperactivity disorder. PLoS ONE 6:e14618. Available at: https://doi.org/10.1371/journal.pone.0014618.Google Scholar
Gonon, F., Konsman, J.-P., Cohen, D. & Boraud, T. (2012) Why most biomedical findings echoed by newspapers turn out to be false: The case of attention deficit hyperactivity disorder. PLoS ONE 7:e44275. Available at: https://doi.org/10.1371/journal.pone.0044275.Google Scholar
Habibzadeh, F. (2013) Common statistical mistakes in manuscripts submitted to biomedical journals. European Science Editing 39:9294.Google Scholar
Haenel, H. (1961) Some rules in the ecology of the intestinal microflora of man. Journal of Applied Bacteriology 24:242–51. Available at: https://doi.org/10.1111/j.1365-2672.1961.tb00260.x.Google Scholar
Hanage, W. P. (2014) Microbiology: Microbiome science needs a healthy dose of scepticism. Nature 512:247–48. Available at: https://doi.org/10.1038/512247a.Google Scholar
Handelsman, J. (2004) Metagenomics: Application of genomics to uncultured microorganisms. Microbiology and Molecular Biology Reviews 68:669–85. Available at: https://doi.org/10.1128/MMBR.68.4.669-685.2004.Google Scholar
Handelsman, J., Rondon, M. R., Brady, S. F., Clardy, J. & Goodman, R. M. (1998) Molecular biological access to the chemistry of unknown soil microbes: A new frontier for natural products. Chemistry & Biology 5:R24549. Available at: https://doi.org/10.1016/S1074-5521(98)90108-9.Google Scholar
Hart, B. L. (1988) Biological basis of the behavior of sick animals. Neuroscience and Biobehavioral Reviews 12:123–37. Available at: http://www.ncbi.nlm.nih.gov/pubmed/3050629.Google Scholar
Hemmings, S. M. J., Malan-Müller, S., van den Heuvel, L. L., Demmitt, B. A., Stanislawski, M. A., Smith, D. G., Bohr, A. D., Stamper, C. E., Hyde, E. R., Morton, J. T., Marotz, C. A., Siebler, P. H., Braspenning, M., Van Criekinge, W., Hoisington, A. J., Brenner, L. A., Postolache, T. T., McQueen, M. B., Krauter, K. S., Knight, R., Seedat, S. & Lowry, C. A. (2017) The microbiome in posttraumatic stress disorder and trauma-exposed controls: An exploratory study. Psychosomatic Medicine 79:936–46. Available at: https://doi.org/10.1097/PSY.0000000000000512.Google Scholar
Hill, C., Guarner, F., Reid, G., Gibson, G. R., Merenstein, D. J., Pot, B., Morelli, L., Canani, R. B., Flint, H. J., Salminen, S., Calder, P. C. & Sanders, M. E. (2014) The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nature Reviews Gastroenterology & Hepatology 11:506–14. Available at: https://doi.org/10.1038/nrgastro.2014.66.Google Scholar
Hodoval, L. F., Morris, E. L., Crawley, G. J. & Beisel, W. R. (1968) Pathogenesis of lethal shock after intravenous staphylococcal enterotoxin B in monkeys. Applied Microbiology 16:187–92. Available at: http://www.ncbi.nlm.nih.gov/pubmed/4967067.Google Scholar
Homberg, J. R. (2013) Measuring behaviour in rodents: Towards translational neuropsychiatric research. Behavioural Brain Research 236:295306. Available at: https://doi.org/10.1016/j.bbr.2012.09.005.Google Scholar
Hooks, K. B. & O'Malley, M. A. (2017) Dysbiosis and its discontents. mBio 8:e01492-17. Available at: https://doi.org/10.1128/mBio.01492-17.Google Scholar
Hsiao, E. Y., McBride, S. W., Hsien, S., Sharon, G., Hyde, E. R., McCue, T., Codelli, J. A., Chow, J., Reisman, S. E., Petrosino, J. F., Patterson, P. H. & Mazmanian, S. K. (2013) Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 155:1451–63. Available at: https://doi.org/10.1016/j.cell.2013.11.024.Google Scholar
Huang, R., Wang, K. & Hu, J. (2016) Effect of probiotics on depression: A systematic review and meta-analysis of randomized controlled trials. Nutrients 8:483. Available at: https://doi.org/10.3390/nu8080483.Google Scholar
Human Microbiome Project Consortium (2012) Structure, function and diversity of the healthy human microbiome. Nature 486(7402):207–14. Available at: https://doi.org/10.1038/nature11234.Google Scholar
Jabr, F. (2017) Probiotics are no panacea. Scientific American 317:2627. Available at: https://doi.org/10.1038/scientificamerican0717-26.Google Scholar
Jeppsson, B., James, J. H., Hummel, R. P., Brenner, W., West, R. & Fischer, J. E. (1983) Increased blood-brain transport of tryptophan after portacaval anastomoses in germ-free rats. Metabolism: Clinical and Experimental 32:48. Available at: https://doi.org/10.1016/0026-0495(83)90147-6.Google Scholar
Jiang, H., Ling, Z., Zhang, Y., Mao, H., Ma, Z., Yin, Y., Wang, W., Tang, W., Tan, Z., Shi, J., Li, L. & Ruan, B. (2015) Altered fecal microbiota composition in patients with major depressive disorder. Brain, Behavior, and Immunity 48:186–94. Available at: https://doi.org/10.1016/j.bbi.2015.03.016.Google Scholar
Johnson, K. V.-A. & Foster, K. R. (2018) Why does the microbiome affect behaviour? Nature Reviews Microbiology 16:647–55. Available at: https://doi.org/10.1038/s41579-018-0014-3.Google Scholar
Kelly, J. R., Allen, A. P., Temko, A., Hutch, W., Kennedy, P. J., Farid, N., Murphy, E., Boylan, G., Bienenstock, J., Cryan, J. F., Clarke, G. & Dinan, T. G. (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 61:5059. Available at: https://doi.org/10.1016/j.bbi.2016.11.018.Google Scholar
Kim, D., Hofstaedter, C. E., Zhao, C., Mattei, L., Tanes, C., Clarke, E., Lauder, A., Sherrill-Mix, S., Chehoud, C., Kelsen, J., Conrad, M., Collman, R. G., Baldassano, R., Bushman, F. D. & Bittinger, K. (2017a) Optimizing methods and dodging pitfalls in microbiome research. Microbiome 5:52. Available at: https://doi.org/10.1186/s40168-017-0267-5.Google Scholar
Kirk, R. G. (2012) “Life in a germ-free world”: Isolating life from the laboratory animal to the bubble boy. Bulletin of the History of Medicine 86:237–75. Available at: https://doi.org/10.1353/bhm.2012.0028.Google Scholar
Kleiman, S. C., Watson, H. J., Bulik-Sullivan, E. C., Huh, E. Y., Tarantino, L. M., Bulik, C. M. & Carroll, I. M. (2015) The intestinal microbiota in acute anorexia nervosa and during renourishment: Relationship to depression, anxiety, and eating disorder psychopathology. Psychosomatic Medicine 77:969–81. Available at: https://doi.org/10.1097/PSY.0000000000000247.Google Scholar
Knight, R., Vrbanac, A., Taylor, B. C., Aksenov, A., Callewaert, C., Debelius, J., Gonzalez, A., Kosciolek, T., McCall, L.-I., McDonald, D., Melnik, A. V., Morton, J. T., Navas, J., Quinn, R. A., Sanders, J. G., Swafford, A. D., Thompson, L. R., Tripathi, A., Xu, Z. Z., Zaneveld, J. R., Zhu, Q., Caporaso, J. G. & Dorrestein, P. C. (2018) Best practices for analysing microbiomes. Nature Reviews Microbiology 16(7):410–22. Available at: https://doi.org/10.1038/s41579-018-0029-9.Google Scholar
Kristensen, N. B., Bryrup, T., Allin, K. H., Nielsen, T., Hansen, T. H. & Pedersen, O. (2016) Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: A systematic review of randomized controlled trials. Genome Medicine 8(1):52. Available at: https://doi.org/10.1186/s13073-016-0300-5.Google Scholar
Leclercq, S., Matamoros, S., Cani, P. D., Neyrinck, A. M., Jamar, F., Stärkel, P., Windey, K., Tremaroli, V., Bäckhed, F., Verbeke, K., de Timary, P. & Delzenne, N. M. (2014) Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of alcohol-dependence severity. Proceedings of the National Academy of Sciences USA 111:E448593. https://doi.org/10.1073/pnas.1415174111.Google Scholar
Lepage, P., Leclerc, M. C., Joossens, M., Mondot, S., Blottière, H. M., Raes, J., Ehrlich, D. & Doré, J. (2013) A metagenomic insight into our gut's microbiome. Gut 62:146–58. Available at: https://doi.org/10.1136/gutjnl-2011-301805.Google Scholar
Liu, R. T. (2017) The microbiome as a novel paradigm in studying stress and mental health. American Psychologist 72(7):655–67. Available at: https://doi.org/10.1037/amp0000058.Google Scholar
Lyte, M. (1993) The role of microbial endocrinology in infectious disease. Journal of Endocrinology 137:343–45. Available at: https://doi.org/10.1677/joe.0.1370343.Google Scholar
Lyte, M. (2011) Probiotics function mechanistically as delivery vehicles for neuroactive compounds: Microbial endocrinology in the design and use of probiotics. BioEssays 33:574–81. Available at: https://doi.org/10.1002/bies.201100024.Google Scholar
Lyte, M. & Ernst, S. (1992) Catecholamine induced growth of gram negative bacteria. Life Sciences 50:203–12. Available at: http://www.ncbi.nlm.nih.gov/pubmed/1731173.Google Scholar
Lyte, M., Varcoe, J. J. & Bailey, M. T. (1998) Anxiogenic effect of subclinical bacterial infection in mice in the absence of overt immune activation. Physiology & Behavior 65:6368. Available at: https://doi.org/10.1016/S0031-9384(98)00145-0.Google Scholar
Magnusson, K. R., Hauck, L., Jeffrey, B. M., Elias, V., Humphrey, A., Nath, R., Perrone, A. & Bermudez, L. E. (2015) Relationships between diet-related changes in the gut microbiome and cognitive flexibility. Neuroscience 300:128–40. Available at: https://doi.org/10.1016/j.neuroscience.2015.05.016.Google Scholar
Mayer, E. A., Knight, R., Mazmanian, S. K., Cryan, J. F. & Tillisch, K. (2014) Gut microbes and the brain: Paradigm shift in neuroscience. Journal of Neuroscience 34(46):15490–96. Available at: https://doi.org/10.1523/JNEUROSCI.3299-14.2014.Google Scholar
Mayer, E. A., Tillisch, K. & Gupta, A. (2015) Gut/brain axis and the microbiota. Journal of Clinical Investigation 125:926–38. Available at: https://doi.org/10.1172/JCI76304.Google Scholar
Mazmanian, S. K., Round, J. L. & Kasper, D. L. (2008) A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 453:620–25. Available at: https://doi.org/10.1038/nature07008.Google Scholar
McKean, J., Naug, H., Nikbakht, E., Amiet, B. & Colson, N. (2017) Probiotics and subclinical psychological symptoms in healthy participants: A systematic review and meta-analysis. Journal of Alternative and Complementary Medicine 23:249–58. Available at: https://doi.org/10.1089/acm.2016.0023.Google Scholar
McNulty, N. P., Yatsunenko, T., Hsiao, A., Faith, J. J., Muegge, B. D., Goodman, A. L., Henrissat, B., Oozeer, R., Cools-Portier, S., Gobert, G., Chervaux, C., Knights, D., Lozupone, C. A., Knight, R., Duncan, A. E., Bain, J. R., Muehlbauer, M. J., Newgard, C. B., Heath, A. C. & Gordon, J. I. (2011) The impact of a consortium of fermented milk strains on the gut microbiome of gnotobiotic mice and monozygotic twins. Science Translational Medicine 3:106r a106. https://doi.org/10.1126/scitranslmed.3002701.Google Scholar
Messaoudi, M., Lalonde, R., Violle, N., Javelot, H., Desor, D., Nejdi, A., Bisson, J.-F., Rougeot, C., Pichelin, M., Cazaubiel, M. & Cazaubiel, J.-M. (2011) Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. British Journal of Nutrition 105:755–64. Available at: https://doi.org/10.1017/S0007114510004319.Google Scholar
Metchnikoff, É. (1908) Études sur la flore intestinale. Putréfaction intestinale. Annales de l'Institut Pasteur 22:930–55.Google Scholar
Molendijk, M. L. & de Kloet, E. R. (2015) Immobility in the forced swim test is adaptive and does not reflect depression. Psychoneuroendocrinology 62:389–91. Available at: https://doi.org/10.1016/j.psyneuen.2015.08.028.Google Scholar
Momozawa, Y., Deffontaine, V., Louis, E. & Medrano, J. F. (2011) Characterization of bacteria in biopsies of colon and stools by high throughput sequencing of the V2 region of bacterial 16S rRNA gene in human. PLoS ONE 6:e16952. Available at: https://doi.org/10.1371/journal.pone.0016952.Google Scholar
Moran, N. A. & Sloan, D. B. (2015) The hologenome concept: Helpful or hollow? PLoS Biology 13:e1002311. Available at: https://doi.org/10.1371/journal.pbio.1002311.Google Scholar
Mormède, P., Andanson, S., Aupérin, B., Beerda, B., Guémené, D., Malmkvist, J., Manteca, X., Manteuffel, G., Prunet, P., van Reenen, C. G., Richard, S. & Veissier, I. (2007) Exploration of the hypothalamic-pituitary-adrenal function as a tool to evaluate animal welfare. Physiology & Behavior 92:317–39. Available at: https://doi.org/10.1016/j.physbeh.2006.12.003.Google Scholar
Neufeld, K., Kang, N., Bienenstock, J. & Foster, J. A. (2011a) Effects of intestinal microbiota on anxiety-like behavior. Communicative & Integrative Biology 4:492–94. Available at: https://doi.org/10.4161/cib.15702.Google Scholar
Neufeld, K., Kang, N., Bienenstock, J. & Foster, J. A. (2011b) Reduced anxiety-like behavior and central neurochemical change in germ-free mice. Neurogastroenterology and Motility 23:255–64, e119. Available at: https://doi.org/10.1111/j.1365-2982.2010.01620.x.Google Scholar
Ng, Q. X., Peters, C., Ho, C. Y. X., Lim, D. Y. & Yeo, W.-S. (2017) A meta-analysis of the use of probiotics to alleviate depressive symptoms. Journal of Affective Disorders 228:1319. Available at: https://doi.org/10.1016/j.jad.2017.11.063.Google Scholar
Nguyen, T. L. A., Vieira-Silva, S., Liston, A. & Raes, J. (2015) How informative is the mouse for human gut microbiota research? Disease Models & Mechanisms 8:116. Available at: https://doi.org/10.1242/dmm.017400.Google Scholar
Nishino, R., Mikami, K., Takahashi, H., Tomonaga, S., Furuse, M., Hiramoto, T., Aiba, Y., Koga, Y. & Sudo, N. (2013) Commensal microbiota modulate murine behaviors in a strictly contamination-free environment confirmed by culture-based methods. Neurogastroenterology and Motility 25:521–e371. Available at: https://doi.org/10.1111/nmo.12110.Google Scholar
Ohland, C. L., Kish, L., Bell, H., Thiesen, A., Hotte, N., Pankiv, E. & Madsen, K. L. (2013) Effects of Lactobacillus helveticus on murine behavior are dependent on diet and genotype and correlate with alterations in the gut microbiome. Psychoneuroendocrinology 38:1738–47. Available at: https://doi.org/10.1016/j.psyneuen.2013.02.008.Google Scholar
Olesen, S. W. & Alm, E. J. (2016) Dysbiosis is not an answer. Nature Microbiology 1:16228. Available at: https://doi.org/10.1038/nmicrobiol.2016.228.Google Scholar
Olle, B. (2013) Medicines from microbiota. Nature Biotechnology 31:309–15. Available at: https://doi.org/10.1038/nbt.2548.Google Scholar
O'Mahony, S. M., Clarke, G., Dinan, T. G. & Cryan, J. F. (2017) Early-life adversity and brain development: Is the microbiome a missing piece of the puzzle? Neuroscience 342:3754. Available at: https://doi.org/10.1016/j.neuroscience.2015.09.068.Google Scholar
O'Mahony, S. M., Marchesi, J. R., Scully, P., Codling, C., Ceolho, A.-M., Quigley, E. M. M., Cryan, J. F. & Dinan, T. G. (2009) Early life stress alters behavior, immunity, and microbiota in rats: Implications for irritable bowel syndrome and psychiatric illnesses. Biological Psychiatry 65:263–67. Available at: https://doi.org/10.1016/j.biopsych.2008.06.026.Google Scholar
O'Malley, M. A. & Skillings, D. J. (2018) Methodological strategies in microbiome research and their explanatory implications. Perspectives on Science 26:239–65. Available at: https://doi.org/10.1162/POSC_a_00274.Google Scholar
Park, A. J., Collins, J., Blennerhassett, P. A., Ghia, J. E., Verdu, E. F., Bercik, P. & Collins, S. M. (2013) Altered colonic function and microbiota profile in a mouse model of chronic depression. Neurogastroenterology and Motility 25:733–e575. Available at: https://doi.org/10.1111/nmo.12153.Google Scholar
Perez-Burgos, A., Wang, B., Mao, Y.-K., Mistry, B., McVey Neufeld, K.-A., Bienenstock, J. & Kunze, W. (2013) Psychoactive bacteria Lactobacillus rhamnosus (JB-1) elicits rapid frequency facilitation in vagal afferents. American Journal of Physiology. Gastrointestinal and Liver Physiology 304:G21120. https://doi.org/10.1152/ajpgi.00128.2012.Google Scholar
Perez-Muñoz, M. E., Arrieta, M.-C., Ramer-Tait, A. E. & Walter, J. (2017) A critical assessment of the “sterile womb” and “in utero colonization” hypotheses: Implications for research on the pioneer infant microbiome. Microbiome 5:48. Available at: https://doi.org/10.1186/s40168-017-0268-4.Google Scholar
Persky, H., Hamburg, D. A., Basowitz, H., Grinker, R. R., Sabshin, M., Korchin, S. J., Herz, M., Board, F. A. & Heath, H. A. (1958) Relation of emotional responses and changes in plasma hydrocortisone level after stressful interview. AMA Archives of Neurology and Psychiatry 79:434–47. Available at: http://www.ncbi.nlm.nih.gov/pubmed/13519947.Google Scholar
Purves, D., Augustine, G. J., Fitzpatrick, D., Katz, L. C., LaMantia, A.-S., McNamara, J. O. & Williams, S. M., eds. (2001) Neuroscience. Sinauer Associates.Google Scholar
Qualliotine, D., DeChatelet, L. R., McCall, C. E. & Cooper, M. R. (1972) Effect of catecholamines on the bactericidal activity of polymorphonuclear leukocytes. Infection and Immunity 6:211–17. Available at: http://www.ncbi.nlm.nih.gov/pubmed/4564885.Google Scholar
Quigley, E. M. M. (2016) Leaky gut – Concept or clinical entity? Current Opinion in Gastroenterology 32:7479. Available at: https://doi.org/10.1097/MOG.0000000000000243.Google Scholar
Quigley, E. M. M. (2017) Gut microbiome as a clinical tool in gastrointestinal disease management: Are we there yet? Nature Reviews Gastroenterology & Hepatology 14:315–20. Available at: https://doi.org/10.1038/nrgastro.2017.29.Google Scholar
Rao, M. & Gershon, M. D. (2016) The bowel and beyond: The enteric nervous system in neurological disorders. Nature Reviews Gastroenterology & Hepatology 13:517–28. Available at: https://doi.org/10.1038/nrgastro.2016.107.Google Scholar
Renaud, M. & Miget, A. (1930) Rôle favorisant des perturbations locales causées par l'adrénaline sur le développement des infections microbiennes. Comptes Rendus des Séances de la Société de Biologie et de ses Filiales 103:1052–54.Google Scholar
Romijn, A. R., Rucklidge, J. J., Kuijer, R. G. & Frampton, C. (2017) A double-blind, randomized, placebo-controlled trial of Lactobacillus helveticus and Bifidobacterium longum for the symptoms of depression. Australian and New Zealand Journal of Psychiatry 51:810–21. Available at: https://doi.org/10.1177/0004867416686694.Google Scholar
Rosen, C. E. & Palm, N. W. (2017) Functional classification of the gut microbiota: The key to cracking the microbiota composition code. BioEssays 39:1700032. Available at: https://doi.org/10.1002/bies.201700032.Google Scholar
Sampson, T. R., Debelius, J. W., Thron, T., Janssen, S., Shastri, G. G., Ilhan, Z. E., Challis, C., Schretter, C. E., Rocha, S., Gradinaru, V., Chesselet, M.-F., Keshavarzian, A., Shannon, K. M., Krajmalnik-Brown, R., Wittung-Stafshede, P., Knight, R. & Mazmanian, S. K. (2016) Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson's disease. Cell 167:1469–80.e12. Available at: https://doi.org/10.1016/j.cell.2016.11.018.Google Scholar
Sampson, T. R. & Mazmanian, S. K. (2015) Control of brain development, function, and behavior by the microbiome. Cell Host & Microbe 17:565–76. Available at: https://doi.org/10.1016/j.chom.2015.04.011.Google Scholar
Savage, D. C. (2001) Microbial biota of the human intestine: A tribute to some pioneering scientists. Current Issues in Intestinal Microbiology 2:115.Google Scholar
Schloss, P. D. (2018) Identifying and overcoming threats to reproducibility, replicability, robustness, and generalizability in microbiome research. mBio 9:e00525-18. Available at: https://doi.org/10.1128/mBio.00525-18.Google Scholar
Schluter, J. & Foster, K. R. (2012) The evolution of mutualism in gut microbiota via host epithelial selection. PLoS Biology 10:e1001424. Available at: https://doi.org/10.1371/journal.pbio.1001424.Google Scholar
Severance, E. G., Yolken, R. H. & Eaton, W. W. (2016) Autoimmune diseases, gastrointestinal disorders and the microbiome in schizophrenia: More than a gut feeling. Schizophrenia Research 176:2335. Available at: https://doi.org/10.1016/j.schres.2014.06.027.Google Scholar
Shade, A. (2017) Diversity is the question, not the answer. ISME Journal 11:16. Available at: https://doi.org/10.1038/ismej.2016.118.Google Scholar
Shanahan, F. & Quigley, E. M. M. (2014) Manipulation of the microbiota for treatment of IBS and IBD – Challenges and controversies. Gastroenterology 146:1554–63. Available at: https://doi.org/10.1053/j.gastro.2014.01.050.Google Scholar
Sherwin, E., Dinan, T. G. & Cryan, J. F. (2018) Recent developments in understanding the role of the gut microbiota in brain health and disease. Annals of the New York Academy of Sciences 1420:525. Available at: https://doi.org/10.1111/nyas.13416.Google Scholar
Slashinski, M. J., McCurdy, S. A., Achenbaum, L. S., Whitney, S. N. & McGuire, A. L. (2012) “Snake-oil,” “quack medicine,” and “industrially cultured organisms”: Biovalue and the commercialization of human microbiome research. BMC Medical Ethics 13:28. Available at: https://doi.org/10.1186/1472-6939-13-28.Google Scholar
Slykerman, R. F., Hood, F., Wickens, K., Thompson, J. M. D., Barthow, C., Murphy, R., Kang, J., Rowden, J., Stone, P., Crane, J., Stanley, T., Abels, P., Purdie, G., Maude, R., Mitchell, E. A. & Probiotic in Pregnancy Study Group. (2017) Effect of Lactobacillus rhamnosus HN001 in pregnancy on postpartum symptoms of depression and anxiety: A randomised double-blind placebo-controlled trial. EBioMedicine 24:159–65. Available at: https://doi.org/10.1016/j.ebiom.2017.09.013.Google Scholar
Smith, P. A. (2015) Can the bacteria in your gut explain your mood? New York Times, June 23. Available at: https://www.nytimes.com/2015/06/28/magazine/can-the-bacteria-in-your-gut-explain-your-mood.html.Google Scholar
Steenbergen, L., Sellaro, R., van Hemert, S., Bosch, J. A. & Colzato, L. S. (2015) A randomized controlled trial to test the effect of multispecies probiotics on cognitive reactivity to sad mood. Brain, Behavior, and Immunity 48:258–64. Available at: https://doi.org/10.1016/j.bbi.2015.04.003.Google Scholar
Stephenson, M. & Rowatt, E. (1947) The production of acetylcholine by a strain of Lactobacillus plantarum. Journal of General Microbiology 1:279–98. Available at: https://doi.org/10.1099/00221287-1-3-279.Google Scholar
Stilling, R. M., Bordenstein, S. R., Dinan, T. G. & Cryan, J. F. (2014) Friends with social benefits: Host-microbe interactions as a driver of brain evolution and development? Frontiers in Cellular and Infection Microbiology 4:147. Available at: https://doi.org/10.3389/fcimb.2014.00147.Google Scholar
Stilling, R. M., Dinan, T. G. & Cryan, J. F. (2016) The brain's Geppetto – microbes as puppeteers of neural function and behaviour? Journal of Neurovirology 22:1421. Available at: https://doi.org/10.1007/s13365-015-0355-x.Google Scholar
Sudo, N., Chida, Y., Aiba, Y., Sonoda, J., Oyama, N., Yu, X.-N., Kubo, C. & Koga, Y. (2004) Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. Journal of Physiology 558(1):263–75. Available at: https://doi.org/10.1113/jphysiol.2004.063388.Google Scholar
Surana, N. K. & Kasper, D. L. (2017) Moving beyond microbiome-wide associations to causal microbe identification. Nature 552:244–47. https://doi.org/10.1038/nature25019.Google Scholar
Swiergiel, A. H. & Dunn, A. J. (2007) Effects of interleukin-1β and lipopolysaccharide on behavior of mice in the elevated plus-maze and open field tests. Pharmacology, Biochemistry, and Behavior 86:651–59. Available at: https://doi.org/10.1016/j.pbb.2007.02.010.Google Scholar
Sze, M. A. & Schloss, P. D. (2016) Looking for a signal in the noise: Revisiting obesity and the microbiome. mBio 7:e01018-16. Available at: https://doi.org/10.1128/mBio.01018-16.Google Scholar
Thompson, A. (2017) Is gut bacteria linked to autism? Pathogens in the stomach alter the brain's development and may increase the risk of condition. Daily Mail, August 25. Available at: http://www.dailymail.co.uk/health/article-4819730/Does-gut-bacteria-cause-autism-Pathogens-alter-brain.html.Google Scholar
Tillisch, K., Labus, J., Kilpatrick, L., Jiang, Z., Stains, J., Ebrat, B., Guyonnet, D., Legrain-Raspaud, S., Trotin, B., Naliboff, B. & Mayer, E.A. (2013) Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology 144(7):1394–401.Google Scholar
Toda, M., Morimoto, K., Nagasawa, S. & Kitamura, K. (2004) Effect of snack eating on sensitive salivary stress markers cortisol and chromogranin a. Environmental Health and Preventive Medicine 9:27. Available at: https://doi.org/10.1265/ehpm.9.27.Google Scholar
Tsavkelova, E. A., Botvinko, I. V, Kudrin, V. S. & Oleskin, A. V. (2000) Detection of neurotransmitter amines in microorganisms with the use of high-performance liquid chromatography. Doklady Biochemistry: Proceedings of the Academy of Sciences of the USSR, Biochemistry Section 372:115–17. Available at: http://www.ncbi.nlm.nih.gov/pubmed/10935181.Google Scholar
Turnbaugh, P. J., Bäckhed, F., Fulton, L. & Gordon, J. I. (2008) Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host & Microbe 3:213–23. Available at: https://doi.org/10.1016/j.chom.2008.02.015.Google Scholar
Turnbaugh, P. J., Ley, R. E., Mahowald, M. A., Magrini, V., Mardis, E. R. & Gordon, J. I. (2006) An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444:1027–31. Available at: https://doi.org/10.1038/nature05414.Google Scholar
Vidgen, B. & Yasseri, T. (2016) P-values: Misunderstood and misused. Frontiers in Physics 4:6. Available at: https://doi.org/10.3389/fphy.2016.00006.Google Scholar
Wang, H., Lee, I.-S., Braun, C. & Enck, P. (2016) Effect of probiotics on central nervous system functions in animals and humans: A systematic review. Journal of Neurogastroenterology and Motility 22:589605. Available at: https://doi.org/10.5056/jnm16018.Google Scholar
Wang, J., Dourmashkin, J. T., Yun, R. & Leibowitz, S. F. (1999) Rapid changes in hypothalamic neuropeptide y produced by carbohydrate-rich meals that enhance corticosterone and glucose levels. Brain Research 848:124–36. Available at: http://www.ncbi.nlm.nih.gov/pubmed/10612704.Google Scholar
Wang, Y. & Kasper, L. H. (2014) The role of microbiome in central nervous system disorders. Brain, Behavior, and Immunity 38:112. Available at: https://doi.org/10.1016/j.bbi.2013.12.015.Google Scholar
Weiss, S., Van Treuren, W., Lozupone, C., Faust, K., Friedman, J., Deng, Y., Xia, L. C., Xu, Z. Z., Ursell, L., Alm, E. J., Birmingham, A., Cram, J. A., Fuhrman, J. A., Raes, J., Sun, F., Zhou, J. & Knight, R. (2016) Correlation detection strategies in microbial data sets vary widely in sensitivity and precision. ISME Journal 10:1669–81. Available at: https://doi.org/10.1038/ismej.2015.235.Google Scholar
Worth, A. R., Lymbery, A. J. & Thompson, R. C. A. (2013) Adaptive host manipulation by Toxoplasma gondii: Fact or fiction? Trends in Parasitology 29:150–55. Available at: https://doi.org/10.1016/j.pt.2013.01.004.Google Scholar
Zaneveld, J. R., McMinds, R. & Vega Thurber, R. (2017) Stress and stability: Applying the Anna Karenina principle to animal microbiomes. Nature Microbiology 2:17121. Available at: https://doi.org/10.1038/nmicrobiol.2017.121.Google Scholar
Zeiss, C. J. & Johnson, L. K. (2017) Bridging the gap between reproducibility and translation: Data resources and approaches. ILAR Journal 58:13. Available at: https://doi.org/10.1093/ilar/ilx017.Google Scholar
Zimmer, C. (2014) Our microbiome may be looking out for itself. New York Times, August 14. Available at: https://www.nytimes.com/2014/08/14/science/our-microbiome-may-be-looking-out-for-itself.html.Google Scholar
Zmora, N., Zilberman-Schapira, G., Suez, J., Mor, U., Dori-Bachash, M., Bashiardes, S., Kotler, E., Zur, M., Regev-Lehavi, D., Brik, R. B., Federici, S., Cohen, Y., Linevsky, R., Rothschild, D., Moor, A. E., Ben-Moshe, S., Harmelin, A., Itzkovitz, S., Maharshak, N., Shibolet, O., Shapiro, H., Pevsner-Fischer, M., Sharon, I., Halpern, Z., Segal, E. & Elinav, E. (2018) Personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features. Cell 174(6):1388–405. Available at: https://doi.org/10.1016/j.cell.2018.08.041.Google Scholar
Figure 0

Table 1. The 25 most cited papers in MGB researcha

Supplementary material: File

Hooks et al. supplementary material

Hooks et al. supplementary material 1

Download Hooks et al. supplementary material(File)
File 549 KB