Abstract
Although the gut and brain are separate organs, they communicate with each other via trillions of intestinal bacteria that collectively make up one’s gut microbiome. Findings from both humans and animals support a critical role of gut microbes in regulating brain function, mood, and behavior. Gut bacteria influence neural circuits that are notably affected in addiction-related behaviors. These include circuits involved in stress, reward, and motivation, with substance use influencing gut microbial abnormalities, suggesting significant gut-brain interactions in drug addiction. Given the overwhelming rates of opioid overdose deaths driven by abuse and addiction, it is essential to characterize mechanisms mediating the abuse potential of opioids. We discuss in this review the role of gut microbiota in factors that influence opioid addiction, including incentive salience, reward, tolerance, withdrawal, stress, and compromised executive function. We present clinical and preclinical evidence supporting a bidirectional relationship between gut microbiota and opioid-related behaviors by highlighting the effects of opioid use on gut bacteria, and the effects of gut bacteria on behavioral responses to opioids. Further, we discuss possible mechanisms of this gut-brain communication influencing opioid use. By clarifying the relationship between the gut microbiome and opioid-related behaviors, we improve understanding on mechanisms mediating reward-, motivation-, and stress-related behaviors and disorders, which may contribute to the development of effective, targeted therapeutic interventions in opioid dependence and addiction.
Keywords: Gut-Brain Axis, Motivation, Bacteria, Dopamine, Stress, Reward, Mood, Addiction
Introduction
A growing field in human health is the gut-brain connection, a notion that our gut health is directly related to our emotional health. Although the gut and brain are separate organs, they are connected physically via the vagus nerve, and biochemically via neurotransmitters, gut hormones, microbial metabolites, and the immune system (Dinan and Cryan, 2012). Bidirectional communication between the central nervous system (brain) and enteric nervous system (gut) occurs through the gut-brain axis, which is maintained by the trillions of intestinal bacteria that collectively make up one’s gut microbiome.
An abundance of evidence in both humans and animals supports an essential role of gut microbiota in regulating brain function, mood, stress, and behavioral responses to rewards, including food and drugs of abuse (Collins and Bercik, 2009; Diaz Heijtz et al., 2011; Clarke et al., 2013; Selkrig et al., 2014; Cussotto et al., 2018). Gut bacteria are heavily impacted by ‟diseased” states, as abnormalities in gut microbial communities, or dysbiosis, are observed in patients diagnosed with substance use disorder, depression, anxiety, Parkinson’s disease, autism, and/or schizophrenia (Schroeder and Bäckhed, 2016). Disruption to a hos’s normal microbiota can lead to exaggerated stress responses and depressive symptoms (Sudo et al., 2004; Ait-Belgnaoui et al., 2014; Tarr et al., 2015; Kelly et al., 2016; Leclercq et al. 2016; Rea et al., 2016), further supporting the bidirectional relationship between the gut and brain. Given the significance of gut bacteria in obesity, stress, and motivated behaviors, as well as the extensive overlap in neuroadaptations between overeating and drug abuse (Volkow et al., 2017) and the key roles of stress and reward processing in the development of addiction, we review available literature to support the hypothesis that gut-brain communication is necessary in the development and perpetuation of drug addiction.
Addiction is a chronic brain disorder that alters circuitry involved in reward, stress, learning, memory, and motivation. The development of drug addiction is driven by exaggerated incentive salience, reward deficits, stress surfeits, and compromised executive function in three distinct stages, namely binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation (craving) (Koob and Volkow, 2016). Opioid addiction is the leading cause of drug overdose in the United States and is characterized by a compulsion to seek and voluntarily take opioids owing to their reinforcing effects and an impaired ability to control intake despite physical or psychological harm (Scholl et al., 2018). Due to the rapid increase in numbers of opioid-related disabilities and deaths throughout the world (Martins et al., 2015), this review is focused on gut-brain interactions specifically in opioid use.
The endogenous opioid system regulates pain relief, reward processing, emotion, stress, and autonomic control, and consists of mu, delta, and kappa receptors (Benarroch, 2012). Opioid receptors are distributed widely throughout the brain, periphery, and gut (De Schepper et al., 2004), and are activated endogenously by enkephalins, dynorphins, endorphins, and endomorphins, as well as exogenously by opioids (e.g., heroin, morphine, oxycodone, fentanyl). Opioids exert their primary clinical effects on mu opioid receptors to reduce pain perception. A link between gut health and opioid intake is evident in the ability of opioids to significantly impact gastrointestinal function (i.e. opioid-induced constipation). Indeed, opioid use is associated with gut dysbiosis in both humans (Vincent et al., 2016; Wang and Roy, 2016; Zhernakova et al., 2016; Acharya et al., 2017; Xu et al., 2017; Barengolts et al., 2018) and animals (Meng et al., 2013; Meng et al., 2015; Banerjee et al., 2016; Kang et al., 2017; Lee et al., 2018; Wang et al., 2018). Preclinical studies have also shown an important role of the gut microbiome in drug reward (Kiraly et al., 2016; Lee et al., 2018) and the development of opioid tolerance (Kang et al., 2017). In this review, we present available literature assessing the bidirectional role of the gut-brain axis in addiction- and opioid-related behaviors, including stress, reward, incentive salience, mood disruption, tolerance, dependence, withdrawal, and antinociception, as well as propose possible mechanisms of gut-brain interactions in opioid use.
With the prevailing opioid crisis and escalating numbers of opioid-related deaths worldwide driven by addiction, it is critical to evaluate mechanisms that mediate the abuse potential of opioids. By presenting a specific role of the gut-brain axis in opioid use and in factors that influence addiction, we provide a potential therapeutic target integrated with opioid regimens to mitigate abuse and addiction vulnerability. Novel interventions to limit the negative clinical outcomes of opioid use, such as tolerance, dependence, and withdrawal, may reduce the risk of addiction and opioid-related deaths. An improved understanding of how the gut is involved in addiction-related behaviors can also contribute to the development of effective treatment strategies in other disorders with shared characteristics, including depression, anxiety, and chronic pain.
Methods to study the gut microbiome
While human subjects provide valuable translational data, the majority of our understanding of gut-brain interactions in addiction-related behaviors comes from preclinical rodent studies. Animal models allow specific, targeted manipulation of gut microbiota while controlling for factors that are widely variable in humans, including disease comorbidities, opioid dose and duration of use, co-use of other opioid and/or non-opioid drugs, drug history, and genetics. Comparing human microbiomes of substance use disorder versus healthy controls may also reflect differences in lifestyles and diets and are therefore difficult to appoint solely to drug use.
The four primary ways that gut microbiota can be manipulated are with probiotics, prebiotics, antibiotics, and fecal microbiota transplantation (FMT). Probiotics are live, beneficial bacterial strains that do not repopulate on their own so need to be administered daily for benefits to the host. Prebiotics are dietary fibers that are indigestible by the host and undergo bacterial fermentation to stimulate the growth of certain types of bacteria. An alternative to prebiotics is administration of short-chain fatty acids (SCFAs), which are bacterial fermentation byproducts. Depletion of 70-90 percent of gut bacteria in rodents can be achieved with prolonged oral treatment of non-absorbable antibiotics that do not mirror clinical doses (Bercik et al., 2011; Reikvam et al., 2011). The goal of FMT, often known as stool transplantation, fecal transplantation, or fecal bacteriotherapy, is to restore eubiosis by transferring stool from a healthy donor into a recipient with an altered colonic microbiome (Vindigni and Surawicz, 2017). FMT is an effective treatment for recurrent Clostridium difficile infection that has not responded to standard therapy. Potential applications of FMT for intervention in non-gastrointestinal diseases, such as obesity, ischemic stroke, Parkinson’s disease, Alzheimer’s disease, and depression, are also being explored (Kelly et al., 2016; Sampson et al., 2016; Chen et al., 2019; Dutta et al., 2019; Muscogiuri et al., 2019; Sun et al., 2019).
A unique animal model used in microbiome research is the germ-free mouse, in which mice are raised in a sterile environment from birth and remain completely absent of internal or external microbes (Faith et al., 2010). Germ-free mice are usually compared with mice containing known pathogens (conventional or specific pathogen-free) and can be colonized with microbial communities from donor animals or human subjects. Although the germ-free animal model is not entirely clinically relevant, as humans have constant exposure to environmental microbes immediately after birth, its use provides insight into early host development and function. Gut microbial depletion via prolonged oral antibiotic administration bypasses the perinatal developmental period.
As the bulk of intestinal bacteria are excreted in fecal matter, microbial analysis is most commonly achieved from fecal samples. Gut bacterial profiles are analyzed by DNA sequencing of the 16S rRNA gene found in all bacteria. Specific primers can select for the variable regions of the 16S gene to provide a profile of the different bacterial species in a given sample. DNA sequencing of bacterial genes reveals a sample’s bacterial abundance, alpha diversity (i.e. how many different species exist in the sample), and beta diversity (i.e. different species in one sample compared to another sample).
Gut microbiome role in incentive salience
Addiction is a chronic brain disease that is associated with dysregulation of reward and motivation. Incentive salience is a motivational property that when attributed to reward-predicting stimuli, or cues, triggers the approach toward and consumption of a reward (Tindell et al., 2009; Zhang et al., 2009). The development of incentive salience is mediated by the mesolimbic dopamine system and promotes habits that encourage excessive cue-induced drug seeking and self-administration behaviors (Berridge 2012).
A vagal gut-to-brain circuit has been established to play a critical role in reward and motivation (Han et al., 2018), and gut bacteria influence how animals respond to various rewards, such as food and drugs (Korner and Leibel, 2003; Duca et al., 2012; Alcock et al., 2014; Kiraly et al., 2016; de Wouw et al., 2018; Lee et al., 2018; Al-Ghezi et al., 2019). Natural rewards, such as food, sex, and nurturing, are processed by key mesocorticolimbic structures and neurotransmitters, including the ventral tegmental area (VTA), nucleus accumbens, prefrontal cortex, dopamine, serotonin, GABA, glutamate, and endogenous opioids (Russo and Nestler, 2013). Drugs of abuse, including opioids, alcohol, stimulants, and cannabis, are artificial rewards that hijack this same brain system, and repeated drug use induces neurophysiological changes that contribute to addiction (Volkow and Morales, 2015).
Passive exposure to or voluntary consumption of drugs of abuse can induce imbalances in gut microbiota in humans and rodents (Volpe et al., 2014; Wang and Roy, 2016; Ning et al., 2017; Temko et al., 2017; Hillemacher et al., 2018; Hofford et al., 2018). In line with bidirectional gut-brain communication, these microbial imbalances influence brain function and behavior. Gut dysbiosis is associated with decreased levels of serotonin and dopamine, both important reward-related neurotransmitters (Yano et al., 2015), and changes in the gut microbiome are correlated with altered striatal dopamine receptor expression in a model of compulsive alcohol seeking (Jadhav et al., 2018). Additionally, mice with depleted gut microbiota have abnormal behavioral responses to cocaine reward compared to controls (Kiraly et al., 2016; Lee et al., 2018), further highlighting a feedback loop between impaired reward processing and gut dysbiosis.
A growing amount of work has investigated the role of gut bacteria in obesity and overeating, which is relatable to drug addiction, as clinical and preclinical evidence uncover a significant overlap of neuroadaptations in overeating (food addiction) and drug addiction (Volkow et al., 2017). The rewarding properties of food and drugs are necessary for addictive potential, as foods that are highly palatable or drugs that produce significant euphoria promote repeated consumption or intake. Further, the rewarding properties of food or drugs are necessary for attributing positive motivational value to stimuli associated with reward availability and act as powerful incentives of rewardseeking behavior (Di Chiara, 1999). Consistent with food and drug reward sharing similar neural mechanisms, current pharmacological and non-pharmacological (i.e. vagal nerve stimulation) treatments for obesity have also shown efficacy in reducing self-administration and/or rewarding effects of alcohol, cocaine, opiates, and nicotine in rodents (Egecioglu et al., 2013; Graham et al., 2013; Skibicka, 2013; Engel and Jerlhag, 2014; Schmidt et al., 2016; Sirohi et al., 2016; Vallof et al., 2016; Childs et al., 2017; Fortin and Roitman, 2017; Tuesta et al., 2017).
As expected, the reward system is more stimulated by high energy-dense food than low-calorie food (van der Laan et al., 2014). High-fat or high-sucrose diets are associated with altered microbial diversity in mice (Daniel et al., 2014; B. Liu et al., 2018; Magnusson et al., 2015), and these diet-induced microbial changes substantially influence brain function, resulting in reduced synaptic plasticity, increased vulnerability to anxiety-like behavior, impairment in long-term and short-term memory, and disruptions in exploratory behavior (Sharma et al., 2012; Bruce-Keller et al., 2015). Bacterial byproducts that come into contact with gut epithelium stimulate production of gut hormones and neuropeptides, including peptide YY, cholecystokinin, glucagon-like peptide-1, and substance P, which mediate hunger and satiety signaling (Cani et al., 2013; Cani and Knauf, 2016). These findings highlight a critical function of gut bacteria in regulating appetite and feeding behaviors.
Although similar neural pathways influence food and drug reward, bariatric surgery (e.g. gastric bypass) to effectively reduce food intake, increases vulnerability and sensitivity to the reinforcing effects of opioid analgesics (Raebel et al., 2013; Biegler et al., 2016). This may be explained by the transference of one addiction (food) to another, as surgery eliminates excessive eating but does not alter individual predispositions to addictive behaviors (Niego et al., 2007; Pepino et al., 2014). In addition, altered gastrointestinal anatomy may cause changes in pharmacokinetics, as opioids are absorbed in the gastrointestinal tract (Tan et al., 1989; Lotsch et al., 1999).
Gut microbiome role in drug withdrawal/negative affect
Various biological factors likely contribute to increasing chronic opioid use, including higher pain sensitivity and lower pain detection thresholds (Dodet et al., 2013). The shift from acute drug use to addiction may be due to opposing brain circuits that mediate stress and reward, as addiction progresses from initial drug use for reward (i.e. positive reinforcement) to repeated use for distress avoidance (i.e. negative reinforcement). Once an individual adapts to regular use of opioids, a sudden decrease in intake leads to withdrawal, which interestingly yields symptoms opposite of opioid effects, as the brain tries to compensate against homeostatic disruption. The withdrawal/negative affect stage of the addiction cycle is represented by dampened reward via dopamine deficits and increased stress via activation of corticotropin-releasing factor and dynorphin (Koob et al., 2014). This recruitment of the stress system results in an emergence of negative emotional states, including dysphoria, anxiety, irritability, and depression. Baseline stress levels are heightened and exacerbated during withdrawal or extended abstinence, which encourages a cycle of continual drug intake to avoid the dysphoric feelings associated with the negative affect.
Stress, both acute and chronic, plays a key role in mediating an animal’s sensitivity to food and drug reinforcers (Sinha and Jastreboff, 2013; Yau and Potenza, 2013; Koob et al., 2014) and is a primary risk factor in the development of drug abuse and addiction. A substantial amount of evidence supports a bidirectional and causal relationship between gut dysbiosis and stress. Gut microbiota directly modulate general stress responses as well as drug withdrawal-induced anxiety (Xiao et al., 2018). Imbalances in gut microbial communities lead to a heightened activation of the hypothalamic-pituitary-adrenal (HPA) axis stress response (Sudo Nobuyuki et al., 2004; Ait-Belgnaoui et al., 2014; Tarr et al., 2015; Gacias et al., 2016; Leclercq et al., 2016; Rea et al., 2016), and restoring eubiosis via probiotics, SCFAs, and FMT ameliorates stress-related biomarkers and behaviors (Desbonnet et al., 2010; Ait-Belgnaoui et al., 2012; Liang et al., 2013; Pusceddu et al., 2015; Schmidt et al., 2015; Tarr et al., 2015). In addition, stress induces changes in microbiota composition and intestinal barrier function (Söderholm et al., 2002; Gareau et al., 2008). This is perhaps unsurprising given the considerable impact that stress has in aggravating gastrointestinal disorders and symptoms, such as inflammatory bowel disease, irritable bowel syndrome, gastric ulcers, and diarrhea (Klooker et al., 2009; Mayer, 2011; Moloney et al., 2016).
Substance use disorders are also highly comorbid with depression and anxiety, which are both characterized as stress-related mood disorders, in which stress is a major risk factor in its onset, and affected individuals have increased stress sensitivity (Holsboer, 2000; Kendler et al., 2006; Scott et al., 2013; Yang et al., 2015; Ramirez et al., 2016). Impaired gut microbiota is reported in depression (Desbonnet et al., 2010; Jiang et al., 2015; Kelly et al., 2016; Luna and Foster, 2015; Macedo et al., 2017) and anxiety (Neufeld et al., 2011; Luna and Foster, 2015; Tarr et al., 2015), as well as other central nervous system abnormalities and diseases, such as hyperactivity, cognitive deficits, Parkinson’s disease, and schizophrenia (Diaz Heijtz et al., 2011; Schepeijans et al., 2015; Frohlich et al., 2016; Severance and Yolken, 2018). This link between central nervous system disorders and dysbiosis is not simply an association, as transplantation of gut bacteria from humans or animal models with obesity, chronic pain, anxiety, depression, Parkinson’s disease, or schizophrenia produces matching abnormal behaviors in animals (Bravo et al., 2011; Bruce-Keller et al., 2015; Kelly et al., 2016; C. Yang et al., 2019; Zhu et al., 2019). Further, gut bacterial depletion in mice increases depressive-like behavior, alters visceral pain responses, and impairs cognition (O’Mahony et al., 2015; Fröhlich et al., 2016). Similarly in humans, a single treatment course of antibiotics is associated with an increased risk of depression and anxiety that rises with recurrent antibiotic exposure (Lurie et al., 2015). These findings collectively suggest a feedback loop exists between stress, depressive states, and gut dysbiosis, which could underlie the gut microbiome’s role in drug withdrawal/negative affect.
Gut microbiome role in drug anticipation/craving
The preoccupation/anticipation stage is often linked with drug craving and hypothesized to be a key element in relapse. During this stage, the combination of excessive drug cue-induced incentive salience, diminished reward system function, and heightened stress levels promotes pathological drug seeking. Motivational withdrawal syndrome develops when access to a drug is prevented, where the primary focus is to alleviate withdrawal symptoms. According to Pavlovian conditioning analysis, drug tolerance and withdrawal symptoms are both manifestations of conditioned compensatory responses (Siegel and Ramos, 2002). These drug-compensatory responses are proposed to mediate the development of tolerance by counteracting the drug effect when administered in the context of usual drug-administration cues (which may also be interpreted as ‟drug preparation” symptoms). In contrast, if the drug is not administered in the presence of usual cues, the conditioned compensatory responses are not attenuated by drug effect and thus achieve full expression, increasing the risk of drug overdose. The essential role of gut microbiota in learning, memory, and stress highlights the striking impact of the gut in drug anticipation and craving (Sudo et al., 2004; Bravo et al., 2011; Diaz Heijtz et al., 2011; Desbonnet et al., 2015).
The finding that opioid tolerance to analgesia can be transferred via fecal transfer from an opioid-dependent mouse model into opioid-naive mice reveals that opioid exposure produces changes in gut bacteria that contribute to the development of tolerance to the pain-relieving effects of opioids (Kang et al., 2017; Lee et al., 2018; Yang et al., 2019). Gut microbiota diversity is in fact altered with chronic opioid use in humans and mice, and these changes alter neuronal tolerance in extrinsic sensory afferents (Akbarali and Dewey, 2017). In addition, the rate of tolerance to the analgesic effects of morphine is exaggerated in the presence of colonic inflammation (Komla et al., 2019).
While tolerance to the analgesic and rewarding properties of opioids develops rapidly, the gastrointestinal-related side effects, including pain, nausea, and constipation, remain consistent and often worsen with chronic opioid exposure (Akbarali and Dewey, 2017). One consideration is that gastrointestinal distress is one presentation of a conditioned compensatory response to opioid intake. As an example, diarrhea and/or vomiting are commonly observed signs of withdrawal, and administration of opioids alleviates these symptoms by inducing constipation and reduced gut motility. Gut dysbiosis may act as an interoceptive cue to opioid administration, similar to external cues, and elicit conditioned responses that mediate drug tolerance (Razran, 1961). Consistent with this hypothesis, tolerance can be prevented with a peripheral mu-opioid receptor antagonist, which supports a peripheral or gut mechanism mediating opioid tolerance (Komla et al., 2019). Furthermore, the insula integrates interoceptive states into emotions and conscious feelings, and its reactivity has been suggested to serve as a biomarker to predict relapse in humans (Naqvi and Bechara, 2009; Janes et al., 2010).
Possible mechanisms of the gut microbiome affecting opioid use behavior
Neuroinflammation
Gut-brain communication occurs vastly through immune pathways. Gut bacteria control the differentiation and function of immune cells in the brain, periphery, and intestines (Erny et al., 2015; Matcovitch-Natan et al., 2016; Rooks and Garrett, 2016), and a healthy intestinal lining forms a tight barrier to control what gets absorbed into the bloodstream. It is therefore feasible to expect that perturbed gut bacteria and/or structure prompt immune dysfunction by triggering inflammation throughout the brain and body.
Intestinal barrier integrity can be threatened by a Western-style diet, certain medications, stress, and autoimmune conditions (Gareau et al., 2007; Stewart et al., 2017), which cause the tight junctions in the large intestine to open up (i.e. leaky gut) and allow bacteria and their toxins to get through, eliciting a systemic inflammatory response. Neuroinflammation is characterized by increased microglial activation and/or malformed microglial morphology. Gut, brain, and systemic inflammation are seen in acute and chronic stress and mood disorders (Maes et al., 2012; Wohleb and Delpech, 2017), as well as in opioid-dependent states and particularly in states of withdrawal (Taylor et al., 2015; Lee et al., 2018). Neuroinflammation disrupts the function and projections of dopaminergic neurons within VTA, leading to decreased mesolimbic dopaminergic activity and dysregulated reward, which is a shared characteristic of chronic pain, depression, and opioid addiction (Taylor et al., 2015; Cahill and Taylor, 2017). Vulnerability to the negative effects of opioids may be heightened by inflammation, which can develop from chronic opioid use, creating a vicious cycle.
Microbial metabolites
Gut bacteria help break down certain nutrients, which can be further metabolized by host cells. Several of these products, short-chain fatty acids (SCFAs, e.g. butyric acid, propionic acid, and acetic acid), are associated with neural function. Gut bacteria also produce tryptophan, serotonin, dopamine, and GABA, which play important roles in the brain as neurotransmitters or their precursors (Lyte, 2011; Thomas et al., 2012; Wall et al., 2014; Sudo, 2019).
SCFAs are considered to be beneficial to the host due to their anti-inflammatory effects and epigenetics regulation (Tsankova et al., 2007; Kim et al., 2014; Emy et al., 2015; Stilling et al., 2016). SCFAs are able to influence memory and learning processes in the brain and alleviate stress (Chambers et al., 2015; J. Liu et al., 2015; Byrne et al., 2016; Amoldussen et al., 2017; Burokas et al., 2017; de Wouw et al., 2018; Garcez et al., 2018). Additionally, administration of SCFAs normalizes microglial abnormalities in germ-free mice (Erny et al., 2015) and reverses the enhanced reward sensitivity to cocaine seen in gut bacteria-depleted mice (Kiraly et al., 2016). Opioid-related behaviors may be due in part to reduced bacterial metabolism, which can be repleted with SCFA supplementation.
Serotonin (5-HT)
Microbiota can regulate 5-hydroxytryptamine (5-HT) synthesis in the gut, which is important given that dysfunctional 5-HT signaling may underlie symptoms of gastrointestinal and mood disorders (Yano et al., 2015). Peripherally, 5-HT is involved in pain perception and regulation of gut secretion and motility, and centrally, 5-HT signaling pathways are implicated in regulating mood and cognition (Gershon and Tack, 2007; O’Mahony et al., 2015). Gut bacteria may play a crucial role in tryptophan availability and metabolism to consequently impact central 5-HT concentrations (de Wouw et al., 2019). The relationship between gut bacteria, serotonin synthesis and signaling, and mood is imperative to note as a consideration in addiction and motivated behaviors.
Microbial diversity and presence of specific species
Dysbiosis is characterized by imbalances in bacterial species, which can be measured by total abundance, species ratios, alpha diversity, and/or the presence or absence of a specific species. An imbalance in the body’s normal gut microbiota disrupts immunity and nutrition and leads to relative overgrowth of bacteria, which can progress into a secondary infection, such as the pathogenic Clostridium difficile. Gut microbiomes with high diversity are posited to be more beneficial to host health than low diversity microbiomes, as many different species exist in low numbers in a high diversity environment and expend more resources competing with other bacteria rather than manipulating the host (Alcock et al., 2014).
Specific bacterial species may exert immunomodulatory effects on the central nervous system. Lactobacillus reuteri (L. reuteri) decreases anxiety-like behavior and stress-induced increase of corticosterone in mice, and alters mRNA expression of both GABA-A and GABA-B receptors in the central nervous system (Bravo et al., 2011). Vagotomy in these animals prevents the anxiolytic and antidepressant effects of L. reuteri, which indicates that parasympathetic innervation is necessary for L. reuteri to participate in the microbiota-brain interaction. Further, many species of Lactobacillus and Bifidobacterium produce GABA; Candida, Escherichia, and Enterococcus produce serotonin; and some Bacillus species produce dopamine (Lyte, 2011; Barrett et al., 2012). Additional research on bacterial species differences in substance use disorder is necessary to understand the functions of specific species and for a precise therapeutic intervention in opioid use.
Conclusion
Few preclinical studies have directly examined the role of gut afferents and bacteria on addiction-like behavior (Kiraly et al., 2016; Han et al., 2018; Lee et al., 2018). Current perspectives focus on dysfunctional reward processing, stress, and mood disorders as risk factors, characteristics, and/or co-morbidities in addiction and their implications with gut dysbiosis. Different pathways mediate the pain-relieving and rewarding effects of opioids, so although an apparent relationship exists between gut bacteria and analgesic tolerance to opioids, further research is needed to evaluate how the gut microbiome modulates opioid reward and reinforcement.
We highlight here the role of gut bacteria in the affected neurocircuitry and behaviors of opioid abuse and addiction, including the stress HPA axis, mesolimbic dopamine system, tolerance, withdrawal, and craving. We also provide support that the gut-brain axis and opioid use share bidirectional communication, as opioid exposure changes the gut microbiome, and manipulation of gut bacteria influences opioid-related behaviors, such as pain tolerance, withdrawal, anhedonia, and drug reward. The impact of gut dysbiosis on impaired reward, enhanced stress, and neuroinflammation, as well as the glaring feedback of these factors on gut health, strongly implicates an important role of the gut-brain axis in opioid use.
Individual differences in gut microbiomes contribute to variations in drug metabolism, which account for the disparities in therapeutic efficacy and side effects between individuals (Zimmermann et al., 2019). An interesting, targeted approach to personalized medicine would be to modulate or supplement the gut microbiota to increase the efficacy of a drug or reduce its adverse effects. Our knowledge of the gut microbiome on obesity has led to clinical trials to evaluate benefits of FMT in non-gastrointestinal disorders, including obesity (Carlucci et al., 2016; Jayasinghe et al., 2016), autism (Kang et al., 2017), multiple sclerosis (Makkawi et al., 2018), and depression (Wortelboer et al., 2019). A further understanding of the role of the gut microbiome in drug addiction and opioid use may offer a novel targeted approach in the treatment of substance use disorders and/or a combined therapy with opioid regimens.
Acknowledgments
Funding: This work was supported by the University of California Irvine (UCI) Microbiome Initiative Pilot Grant Program (SL), UCI School of Medicine start up fund (SL), UCI Institute for Clinical and Translational Sciences (ICTS) Pilot Studies Program (NUT/NCATS) (SL), Helping End Addiction Long-Term (HEAL) Initiative Opioid-Related Pilot Studies Program (ICTS, UCI School of Medicine, Department of Anesthesiology and Perioperative Care) (SL), UCI Department of Emergency Medicine Pilot Award (SL), and UCI Center for the Neurobiology of Learning and Memory Pedagogical Fellowship (MR).
Footnotes
Conflicts of Interest: None
References
- Acharya C, Betrapally N, Gillevet P, Sterling R, Akbarali H, White M, et al. (2017). Chronic opioid use is associated with altered gut microbiota and predicts readmissions in patients with cirrhosis. Alimentary Pharmacology and Therapeutics 45(2):319–331. [DOI] [PubMed] [Google Scholar]
- Ait-Belgnaoui A, Colom A, Braniste V, Ramalho L, Marrot A, Cartier C, et al. (2014). Probiotic gut effect prevents the chronic psychological stress-induced brain activity abnormality in mice. Neurogastroenterology and Motility 26(4):510–520. 10.1111/nmo.12295 [DOI] [PubMed] [Google Scholar]
- Ait-Belgnaoui A, Durand H, Cartier C, Chaumaz G, Eutamene H, Ferrier L, et al. (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—1895. 10.1016/j.psyneuen.2012.03.024 [DOI] [PubMed] [Google Scholar]
- Akbarali HI, Dewey WL (2017). The gut—brain interaction in opioid tolerance. Current Opinion in Pharmacology 37:126–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alcock J, Maley CC, Aktipis CA (2014). Is eating behavior manipulated by the gastrointestinal microbiota? Evolutionary pressures and potential mechanisms. Bioessays 36(10): 940–949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Al-Ghezi ZZ, Busbee PB, Alghetaa H, Nagarkatti PS, Nagarkatti M (2019). Combination of cannabinoids, delta-9-tetrahydrocannabinol (THC) and cannabidiol (CBD), mitigates experimental autoimmune encephalomyelitis (EAE) by altering the gut microbiome. Brain, Behavior, and Immunity 82:25–35. 10.1016/j.bbi.2019.07.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arnoldussen I, Wiesmann M, Pelgrim C, Wielemaker E, van Duyvenvoorde W, Amaral-Santos P, et al. (2017). Butyrate restores HFD-induced adaptations in brain function and metabolism in mid-adult obese mice. International Journal of Obesity 41(6):935. [DOI] [PubMed] [Google Scholar]
- Banerjee S, Sindberg G, Wang F, Meng J, Sharma U, Zhang L, et al. (2016). Opioid-induced gut microbial disruption and bile dysregulation leads to gut barrier compromise and sustained systemic inflammation. Mucosal Immunology 9:1418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barengolts E, Green SJ, Eisenberg Y, Akbar A, Reddivari B, Layden BT, et al. (2018). Gut microbiota varies by opioid use, circulating leptin and oxytocin in African American men with diabetes and high burden of chronic disease. PLOS ONE 13(3):e0194171. 10.1371/journal.pone.0194171 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barrett E, Ross RP, O’Toole PW, Fitzgerald GF, Stanton C (2012). γ-Aminobutyric acid production by culturable bacteria from the human intestine. Journal of Applied Microbiology 113(2):411–417. 10.1111/j.1365-2672.2012.05344.X [DOI] [PubMed] [Google Scholar]
- Benarroch EE (2012). Endogenous opioid systems. Neurology 79(8):807. 10.1212/WNL.0b013e3182662098 [DOI] [PubMed] [Google Scholar]
- Bercik P, Denou E, Collins J, Jackson W, Lu J, Jury J, et al. (2011). The Intestinal Microbiota Affect Central Levels of Brain-Derived Neurotropic Factor and Behavior in Mice. Gastroenterology 141(2):599–609.e3. 10.1053/j.gastro.2011.04.052 [DOI] [PubMed] [Google Scholar]
- Berridge KC (2012). From prediction error to incentive salience: mesolimbic computation of reward motivation. The European Journal of Neuroscience 35(7): 1124–1143. 10.1111/j.1460-9568.2012.07990.X [DOI] [PMC free article] [PubMed] [Google Scholar]
- Biegler JM, Freet CS, Horvath N, Rogers AM, Hajnal A (2016). Increased intravenous morphine self-administration following Roux-en-Y gastric bypass in dietary obese rats. Brain Research Bulletin 123:47–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, et al. (2011). Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proceedings of the National Academy of Sciences of the United States of America 108(38): 16050–16055. 10.1073/pnas.1102999108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bruce-Keller AJ, Salbaum JM, Luo M, Blanchard IVE, Taylor CM, Welsh DA, Berthoud HR (2015). Obese-type gut microbiota induce neurobehavioral changes in the absence of obesity. Biological Psychiatry 77(7):607–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burokas A, Arboleya S, Moloney RD, Peterson VL, Murphy K, Clarke G, et al. (2017). Targeting the microbiota-gut-brain axis: prebiotics have anxiolytic and antidepressant-like effects and reverse the impact of chronic stress in mice. Biological Psychiatry 82(7):472–487. [DOI] [PubMed] [Google Scholar]
- Byrne CS, Chambers ES, Alhabeeb H, Chhina N, Morrison DJ, Preston T, et al. (2016). Increased colonic propionate reduces anticipatory reward responses in the human striatum to high-energy foods. The American Journal of Clinical Nutrition 104(1):5–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cahill CM, Taylor AM (2017). Neuroinflammation-a co-occurring phenomenon linking chronic pain and opioid dependence. Current Opinion in Behavioral Sciences 13:171–177. 10.1016/j.cobeha.2016.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cani PD, Everard A, Duparc T (2013). Gut microbiota, enteroendocrine functions and metabolism. Gastrointestinal Endocrine and Metabolic Diseases 13(6):935–940. 10.1016/j.coph.2013.09.008 [DOI] [PubMed] [Google Scholar]
- Cani PD, Knauf C (2016). How gut microbes talk to organs: The role of endocrine and nervous routes. Molecular Metabolism 5(9):743–752. 10.1016/j.molmet.2016.05.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carlucci C, Petrof EO, Allen-Vercoe E (2016). Fecal Microbiota-based Therapeutics for Recurrent Clostridium difficile Infection, Ulcerative Colitis and Obesity. EBioMedicine 13:37–45. 10.1016/j.ebiom.2016.09.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chambers ES, Viardot A, Psichas A, Morrison DJ, Murphy KG, Zac-Varghese SE, et al. (2015). Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 64(11): 1744–1754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen R, Xu Y, Wu P, Zhou H, Lasanajak Y, Fang Y, et al. (2019). Transplantation of Fecal Microbiota Rich in Short Chain Fatty Acids and Butyric Acid Treat Cerebral Ischemic Stroke by Regulating Gut Microbiota. Pharmacological Research 104403. 10.1016/j.phrs.2019.104403 [DOI] [PubMed] [Google Scholar]
- Childs JE, DeLeon J, Nickel E, Kroener S (2017). Vagus nerve stimulation reduces cocaine seeking and alters plasticity in the extinction network. Learning and Memory 24(1):35–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Clarke G, Grenham S, Scully P, Fitzgerald P, Moloney R, Shanahan F, et al. (2013). The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Molecular Psychiatry 18(6):666. [DOI] [PubMed] [Google Scholar]
- Collins SM, Bercik P (2009). The Relationship Between Intestinal Microbiota and the Central Nervous System in Normal Gastrointestinal Function and Disease. Intestinal Microbes in Health and Disease 136(6):2003–2014. 10.1053/j.gastro.2009.01.075 [DOI] [PubMed] [Google Scholar]
- Cussotto S, Sandhu KV, Dinan TG, Cryan JF (2018). The Neuroendocrinology of the Microbiota-Gut-Brain Axis: A Behavioural Perspective. Frontiers in Neuroendocrinology. 10.1016/j.yfrne.2018.04.002 [DOI] [PubMed] [Google Scholar]
- Daniel H, Gholami AM, Berry D, Desmarchelier C, Hahne H, Loh G, et al. (2014). High-fat diet alters gut microbiota physiology in mice. The ISME Journal 8(2):295–308. 10.1038/ismej.2013.155 [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Schepper HU, Cremonini F, Park MI, Camilleri M (2004). Opioids and the gut: pharmacology and current clinical experience. Neurogastroenterology and Motility 16(4):383–394. 10.1111/j.1365-2982.2004.00513.x [DOI] [PubMed] [Google Scholar]
- de Wouw M, Boehme M, Lyte JM, Wiley N, Strain C, O’Sullivan O, et al. (2018). Short-chain fatty acids: microbial metabolites that alleviate stress-induced brain-gut axis alterations. The Journal of Physiology 596:4923–4944. 10.1113/JP276431 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Desbonnet L, Garrett L, Clarke G, Kiely B, Cryan JF, Dinan TG (2010). Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression. Neuroscience 170(4): 1179–1188. 10.1016/j.neuroscience.2010.08.005 [DOI] [PubMed] [Google Scholar]
- Di Chiara G (1999). Drug addiction as dopamine-dependent associative learning disorder. European Journal of Pharmacology 375(1):13–30. 10.1016/S0014-2999(99)00372-6 [DOI] [PubMed] [Google Scholar]
- Diaz Heijtz R, Wang S, Anuar F, Qian Y, Björkholm B, Samuelsson A, et al. (2011). Normal gut microbiota modulates brain development and behavior. Proceedings of the National Academy of Sciences of the United States of America 108(7):3047–3052. 10.1073/pnas.1010529108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dinan TG, Cryan JF (2012). Regulation of the stress response by the gut microbiota: Implications for psychoneuroendocrinology. Psychoneuroendocrinology 37(9): 1369–1378. 10.1016/j.psyneuen.2012.03.007 [DOI] [PubMed] [Google Scholar]
- Dodet P, Perrot S, Auvergne L, Hajj A, Simoneau G, Declèves X, et al. (2013). Sensory impairment in obese patients? Sensitivity and pain detection thresholds for electrical stimulation after surgery-induced weight loss, and comparison with a nonobese population. The Clinical Journal of Pain 29(1):43–49. [DOI] [PubMed] [Google Scholar]
- Duca FA, Swartz TD, Sakar Y, Covasa M (2012). Increased Oral Detection, but Decreased Intestinal Signaling for Fats in Mice Lacking Gut Microbiota. PLoS ONE 7(6):e39748. 10.1371/journal.pone.0039748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dutta SK, Verma S, Jain V, Surapaneni BK, Vinayek R, Phillips L, Nair PP (2019). Parkinson’s Disease: The Emerging Role of Gut Dysbiosis, Antibiotics, Probiotics, and Fecal Microbiota Transplantation. Journal of Neurogastroenterology and Motility 25(3):363–376. 10.5056/jnml9044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Egecioglu E, Engel JA, Jerlhag E (2013). The Glucagon-Like Peptide 1 Analogue, Exendin-4, Attenuates the Rewarding Properties of Psychostimulant Drugs in Mice. PLoS ONE 8(7):e69010. 10.1371/journal.pone.0069010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Engel JA, Jerlhag E (2014). Role of Appetite-Regulating Peptides in the Pathophysiology of Addiction: Implications for Pharmacotherapy. CNS Drugs 28(10):875–886. 10.1007/s40263-014-0178-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erny D, de Angelis ALH, Jaitin D, Wieghofer P, Staszewski O, David E, et al. (2015). Host microbiota constantly control maturation and function of microglia in the CNS. Nature Neuroscience 18(7):965–977. 10.1038/nn.4030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Faith JJ, Rey FE, O’Donnell D, Karlsson M, McNulty NP, Kallstrom G, et al. (2010). Creating and characterizing communities of human gut microbes in gnotobiotic mice. The ISME Journal 4(9): 1094–1098. 10.1038/ismej.2010.110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fortin SM, Roitman MF (2017). Central GLP-1 receptor activation modulates cocaine-evoked phasic dopamine signaling in the nucleus accumbens core. Proceedings of the SSIB 2016 Annual Meeting 176:17–25. 10.1016/j.physbeh.2017.03.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fröhlich EE, Farzi A, Mayerhofer R, Reichmann F, Jačan A, Wagner B, et al. (2016). Cognitive impairment by antibiotic-induced gut dysbiosis: Analysis of gut microbiota-brain communication. Brain, Behavior, and Immunity 56:140–155. 10.1016/j.bbi.2016.02.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gacias M, Gaspari S, Santos PMG, Tamburini S, Andrade M, Zhang F, et al. (2016). Microbiota-driven transcriptional changes in prefrontal cortex override genetic differences in social behavior. ELife 5:e13442. 10.7554/eLife.13442 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garcez ML, de Carvalho CA, Mina F, Bellettini-Santos T, Schiavo GL, da Silva S, et al. (2018). Sodium butyrate improves memory and modulates the activity of histone deacetylases in aged rats after the administration of d-galactose. Experimental Gerontology 113:209–217. 10.1016/j.exger.2018.10.005 [DOI] [PubMed] [Google Scholar]
- Gareau MG, Jury J, MacQueen G, Sherman PM, Perdue MH (2007). Probiotic treatment of rat pups normalises corticosterone release and ameliorates colonic dysfunction induced by maternal separation. Gut 56(11): 1522–1528. 10.1136/gut.2006.117176 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gershon MD, Tack J (2007). The serotonin signaling system: from basic understanding to drug development for functional GI disorders. Gastroenterology 132(1):397–414. [DOI] [PubMed] [Google Scholar]
- Graham DL, Erreger K, Galli A, Stanwood GD (2013). GLP-1 analog attenuates cocaine reward. Molecular Psychiatry 18(9):961–962. 10.1038/mp.2012.141 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han W, Tellez LA, Perkins MH, Perez IO, Qu T, Ferreira J, et al. (2018). A Neural Circuit for Gut-Induced Reward. Cell 175(3):665–678. 10.1016/j.cell.2018.08.049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hillemacher T, Bachmann O, Kahl KG, Frieling H (2018). Alcohol, microbiome, and their effect on psychiatric disorders. Progress in Neuro-Psychopharmacology and Biological Psychiatry 85:105–115. 10.1016/j.pnpbp.2018.04.015 [DOI] [PubMed] [Google Scholar]
- Hofford RS, Russo SJ, Kiraly DD (2018). Neuroimmune mechanisms of psychostimulant and opioid use disorders. European Journal of Neuroscience. https://doi.org/10.1111/ejn.14143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holsboer F (2000). The Corticosteroid Receptor Hypothesis of Depression. Neuropsychopharmacology 23(5):477–501. 10.1016/S0893-133X(00)00159-7 [DOI] [PubMed] [Google Scholar]
- Jadhav KS, Peterson VL, Halfon O, Ahern G, Fouhy F, Stanton C, et al. (2018). Gut microbiome correlates with altered striatal dopamine receptor expression in a model of compulsive alcohol seeking. Neuropharmacology 141:249–259. 10.1016/j.neuropharm.2018.08.026 [DOI] [PubMed] [Google Scholar]
- Janes AC, Pizzagalli DA, Richardt S, Frederick BB, Chuzi S, Pachas G, et al. (2010). Brain reactivity to smoking cues prior to smoking cessation predicts ability to maintain tobacco abstinence. Biological Psychiatry 67(8):722–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jayasinghe TN, Chiavaroli V, Holland DJ, Cutfield WS, O’Sullivan JM (2016). The New Era of Treatment for Obesity and Metabolic Disorders: Evidence and Expectations for Gut Microbiome Transplantation. Frontiers in Cellular and Infection Microbiology 6:15–15. 10.3389/fcimb.2016.00015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang H, Ling Z, Zhang Y, Mao H, Ma Z, Yin Y, et al. (2015). Altered fecal microbiota composition in patients with major depressive disorder. Brain, Behavior, and Immunity 48:186–194. 10.1016/j.bbi.2015.03.016 [DOI] [PubMed] [Google Scholar]
- Kang DW, Adams JB, Gregory AC, Borody T, Chittick L, Fasano A, et al. (2017). Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome 5(1): 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang M, Mischel RA, Bhave S, Komla E, Cho A, Huang C, et al. (2017). The effect of gut microbiome on tolerance to morphine mediated antinociception in mice. Scientific Reports 7:42658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelly JR, Borre Y, O’ Brien C, Patterson E, El Aidy S, Deane J, et al. (2016). Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat. Journal of Psychiatric Research 82:109–118. 10.1016/j.jpsychires.2016.07.019 [DOI] [PubMed] [Google Scholar]
- Kendler KS, Gatz M, Gardner CO, Pedersen NL (2006). A Swedish National Twin Study of Lifetime Major Depression. American Journal of Psychiatry 163(1): 109–114. 10.1176/appi.ajp.163.1.109 [DOI] [PubMed] [Google Scholar]
- Kim CH, Park J, Kim M (2014). Gut microbiota-derived short-chain Fatty acids, T cells, and inflammation. Immune Network 14(6):277–288. 10.4110/in.2014.14.6.277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kiraly DD, Walker DM, Calipari ES, Labonte B, Issler O, Pena CJ, et al. (2016). Alterations of the Host Microbiome Affect Behavioral Responses to Cocaine. Scientific Reports 6:35455. 10.1038/srep35455 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klooker TK, Braak B, Painter RC, De Rooij SR, Van Elburg RM, Van Den Wijngaard RM, et al. (2009). Exposure to severe wartime conditions in early life is associated with an increased risk of irritable bowel syndrome: a population-based cohort study. The American Journal of Gastroenterology 104(9):2250. [DOI] [PubMed] [Google Scholar]
- Komla E, Stevens DL, Zheng Y, Zhang Y, Dewey WL, Akbarali HI (2019). Experimental colitis enhances the rate of antinociceptive tolerance to morphine via peripheral opioid receptors. Journal of Pharmacology and Experimental Therapeutics jpet. 119.256941. 10.1124/jpet.119.256941 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koob GF, Buck CL, Cohen A, Edwards S, Park PE, Schlosburg JE, et al. (2014). Addiction as a stress surfeit disorder. Neuropharmacology 76:370–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koob GF, Volkow ND (2016). Neurobiology of addiction: a neurocircuitry analysis. The Lancet Psychiatry 3(8):760–773. 10.1016/S2215-0366(16)00104-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Korner J, Leibel RL (2003). To eat or not to eat-how the gut talks to the brain. New England Journal of Medicine 349(10): 926–927. [DOI] [PubMed] [Google Scholar]
- Leclercq S, Forsythe P, Bienenstock J (2016). Posttraumatic Stress Disorder: Does the Gut Microbiome Hold the Key? Canadian Journal of Psychiatry. Revue Canadienne de Psychiatrie 61(4):204–213. 10.1177/0706743716635535 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee K, Vuong HE, Nusbaum DJ, Hsiao EY, Evans CJ, Taylor AMW (2018). The gut microbiota mediates reward and sensory responses associated with regimen-selective morphine dependence. Neuropsychopharmacology 43:2606–2614. 10.1038/s41386-018-0211-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liang NC, Bello N, Moran T (2013). Additive feeding inhibitory and aversive effects of naltrexone and exendin-4 combinations. International Journal of Obesity (2005) 37(2):272–278. 10.1038/ijo.2012.16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu B, Zhang Y, Wang R, An Y, Gao W, Bai L, et al. (2018). Western diet feeding influences gut microbiota profiles in apoE knockout mice. Lipids in Health and Disease 17(1): 159–159. 10.1186/s12944-018-0811-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu J, Sun J, Wang F, Yu X, Ling Z, Li H, et al. (2015). Neuroprotective effects of Clostridium butyricum against vascular dementia in mice via metabolic butyrate. BioMed Research International 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lotsch J, Weiss M, Ahne G, Kobal G, Geisslinger G (1999). Pharmacokinetic modeling of M6G formation after oral administration of morphine in healthy volunteers. Anesthesiology: The Journal of the American Society of Anesthesiologists 90(4): 1026–1038. [DOI] [PubMed] [Google Scholar]
- Luna RA, Foster JA (2015). Gut brain axis: diet microbiota interactions and implications for modulation of anxiety and depression. Food Biotechnology Plant Biotechnology 32:35–41. 10.1016/j.copbio.2014.10.007 [DOI] [PubMed] [Google Scholar]
- Lurie I, Yang YX, Haynes K, Mamtani R, Boursi B (2015). Antibiotic exposure and the risk for depression, anxiety, or psychosis: a nested case-control study. The Journal of Clinical Psychiatry 76(11): 1522–1528. [DOI] [PubMed] [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(8):574–581. 10.1002/bies.201100024 [DOI] [PubMed] [Google Scholar]
- Macedo D, Filho AJMC, Soares de Sousa CN, Quevedo J, Barichello T, Júnior HVN, Freitas de Lucena D (2017). Antidepressants, antimicrobials or both? Gut microbiota dysbiosis in depression and possible implications of the antimicrobial effects of antidepressant drugs for antidepressant effectiveness. Journal of Affective Disorders 208:22–32. 10.1016/j.jad.2016.09.012 [DOI] [PubMed] [Google Scholar]
- Maes M, Berk M, Goehler L, Song C, Anderson G, Gałecki P, Leonard B (2012). Depression and sickness behavior are Janus-faced responses to shared inflammatory pathways. BMC Medicine 10:66–66. 10.1186/1741-7015-10-66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Magnusson K, Hauck L, Jeffrey B, Elias V, Humphrey A, Nath R, et al. (2015). Relationships between diet-related changes in the gut microbiome and cognitive flexibility. Neuroscience 300:128–140. [DOI] [PubMed] [Google Scholar]
- Makkawi S, Camara-Lemarroy C, Metz L (2018). Fecal microbiota transplantation associated with 10 years of stability in a patient with SPMS. Neurology-Neuroimmunology Neuroinflammation 5(4):e459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martins SS, Sampson L, Cerdá M, Galea S (2015). Worldwide Prevalence and Trends in Unintentional Drug Overdose: A Systematic Review of the Literature. American Journal of Public Health 105(11):e29–e49. 10.2105/AJPH.2015.302843 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matcovitch-Natan O, Winter DR, Giladi A, Vargas Aguilar S, Spinrad A, Sarrazin S, et al. (2016). Microglia development follows a stepwise program to regulate brain homeostasis. Science 353(6301):8670. 10.1126/science.aad8670 [DOI] [PubMed] [Google Scholar]
- Mayer EA (2011). Gut feelings: the emerging biology of gut-brain communication. Nature Reviews Neuroscience 12(8):453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gareau MG, Silva MA, Perdue MH (2008). Pathophysiological Mechanisms of Stress-Induced Intestina Damage. Current Molecular Medicine 8(4):274–281. 10.2174/156652408784533760 [DOI] [PubMed] [Google Scholar]
- Meng J, Sindberg GM, Roy S (2015). Disruption of gut homeostasis by opioids accelerates HIV disease progression. Frontiers in Microbiology 6:643. 10.3389/fmicb.2015.00643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meng J, Yu H, Ma J, Wang J, Banerjee S, Charboneau R, et al. (2013). Morphine Induces Bacterial Translocation in Mice by Compromising Intestinal Barrier Function in a TLR-Dependent Manner. PLOS ONE 8(1):e54040. 10.1371/journal.pone.0054040 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moloney RD, Johnson AC, O’Mahony SM, Dinan TG, Greenwood-Van Meerveld B, Cryan JF (2016). Stress and the microbiota-gut-brain axis in visceral pain: relevance to irritable bowel syndrome. CNS Neuroscience and Therapeutics 22(2): 102–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muscogiuri G, Cantone E, Cassarano S, Tuccinardi D, Barrea L, Savastano S, et al. (2019). Gut microbiota: a new path to treat obesity. International Journal of Obesity Supplements 9(1): 10–19. 10.1038/s41367-019-0011-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Naqvi NH, Bechara A (2009). The hidden island of addiction: the insula. Trends in Neurosciences 32(1):56–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neufeld KAM, Kang N, Bienenstock J, Foster JA (2011). Effects of intestinal microbiota on anxiety-like behavior. Communicative and Integrative Biology 4(4):492–494. 10.4161/cib.4.4.15702 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Niego SH, Kofman MD, Weiss JJ, Geliebter A (2007). Binge eating in the bariatric surgery population: a review of the literature. International Journal of Eating Disorders 40(4):349–359. [DOI] [PubMed] [Google Scholar]
- Ning T, Gong X, Xie L, Ma B (2017). Gut microbiota analysis in rats with methamphetamine-induced conditioned place preference. Frontiers in Microbiology 8:1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Mahony SM, Clarke G, Borre Y, Dinan T, Cryan J (2015). Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behavioural Brain Research 277:32–48. [DOI] [PubMed] [Google Scholar]
- Pepino MY, Stein RI, Eagon JC, Klein S (2014). Bariatric surgery-induced weight loss causes remission of food addiction in extreme obesity. Obesity (Silver Spring, Md.) 22(8): 1792–1798. 10.1002/oby.20797 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pusceddu MM, El Aidy S, Crispie F, O’Sullivan O, Cotter P, Stanton C, et al. (2015). N-3 Polyunsaturated Fatty Acids (PUFAs) Reverse the Impact of Early-Life Stress on the Gut Microbiota. PloS One 10(10):e0139721–e0139721. 10.1371/journal.pone.0139721 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raebel MA, Newcomer SR, Reifler LM, Boudreau D, Elliott TE, DeBar L, et al. (2013). Chronic use of opioid medications before and after bariatric surgery. Jama 310(13): 1369–1376. [DOI] [PubMed] [Google Scholar]
- Ramirez K, Fomaguera-Trías J, Sheridan JF (2016). Stress-induced microglia activation and monocyte trafficking to the brain underlie the development of anxiety and depression. In Inflammation-Associated Depression: Evidence, Mechanisms and Implications; 155–172. Springer. [DOI] [PubMed] [Google Scholar]
- Razran G (1961). The observable unconscious and the inferable conscious in current Soviet psychophysiology: Interoceptive conditioning, semantic conditioning, and the orienting reflex. Psychological Review 104:170–193. [PubMed] [Google Scholar]
- Rea K, Dinan TG, Cryan JF (2016). The microbiome: A key regulator of stress and neuroinflammation. Neurobiology of Stress 4:23–33. 10.1016/j.ynstr.2016.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reikvam DH, Erofeev A, Sandvik A, Grcic V, Jahnsen FL, Gaustad P, et al. (2011). Depletion of murine intestinal microbiota: effects on gut mucosa and epithelial gene expression. PloS One 6(3):e17996–e17996. 10.1371/journal.pone.0017996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rooks MG, Garrett WS (2016). Gut microbiota, metabolites and host immunity. Nature Reviews. Immunology 16(6):341–352. 10.1038/nri.2016.42 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Russo SJ, Nestler EJ (2013). The brain reward circuitry in mood disorders. Nature Reviews. Neuroscience 14(9):609–625. 10.1038/nri3381 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, et al. (2016). Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 167(6): 1469–1480.e12. 10.1016/j.cell.2016.11.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scheperjans F, Aho V, Pereira PAB, Koskinen K, Paulin L, Pekkonen E, et al. (2015). Gut microbiota are related to Parkinson’s disease and clinical phenotype. Movement Disorders 30(3):350–358. 10.1002/mds.26069 [DOI] [PubMed] [Google Scholar]
- Schmidt HD, Mietlicki-Baase EG, Ige KY, Maurer JJ, Reiner DJ, Zimmer DJ, et al. (2016). Glucagon-Like Peptide-1 Receptor Activation in the Ventral Tegmental Area Decreases the Reinforcing Efficacy of Cocaine. Neuropsychopharmacology 41(7): 1917–1928. 10.1038/npp.2015.362 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmidt K, Cowen PJ, Harmer CJ, Tzortzis G, Errington S, Burnet PW (2015). Prebiotic intake reduces the waking cortisol response and alters emotional bias in healthy volunteers. Psychopharmacology 232(10): 1793–1801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Scholl L, Seth P, Kariisa M, Wilson N, Baldwin G (2018). Drug and Opioid-Involved Overdose Deaths - United States, 2013-2017. MMWR Morb Mortal Wkly Rep 67:1419–1427. 10.15585/mmwr.mm675152e1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schroeder BO, Bäckhed F (2016). Signals from the gut microbiota to distant organs in physiology and disease. Nature Medicine 22:1079. [DOI] [PubMed] [Google Scholar]
- Scott LV, Clarke G, Dinan TG (2013). The Brain-Gut Axis: A Target for Treating Stress-Related Disorders. In Modern Trends in Psychiatry 28:90–99. 10.1159/000343971 [DOI] [PubMed] [Google Scholar]
- Selkrig J, Wong P, Zhang X, Pettersson S (2014). Metabolic tinkering by the gut microbiome: Implications for brain development and function. Gut Microbes 5(3):369–380. 10.4161/gmic.28681 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Severance EG, Yolken RH (2018). Deciphering microbiome and neuroactive immune gene interactions in schizophrenia. Neurobiology of Disease. 10.1016/j.nbd.2018.11.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sharma S, Zhuang Y, Gomez-Pinilla F (2012). High-fat diet transition reduces brain DHA levels associated with altered brain plasticity and behaviour. Scientific Reports 2:431–431. 10.1038/srep00431 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siegel S, Ramos BMC (2002). Applying laboratory research: Drug anticipation and the treatment of drug addiction. Experimental and Clinical Psychopharmacology 10:162–183. [DOI] [PubMed] [Google Scholar]
- Sinha R, Jastreboff AM (2013). Stress as a common risk factor for obesity and addiction. Biological Psychiatry 73(9):827–835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sirohi S, Schurdak JD, Seeley RJ, Benoit SC, Davis JF (2016). Central and peripheral glucagon-like peptide-1 receptor signaling differentially regulate addictive behaviors. Physiology and Behavior 161:140–144. 10.1016/j.physbeh.2016.04.013 [DOI] [PubMed] [Google Scholar]
- Skibicka KP (2013). The central GLP-1: implications for food and drug reward. Frontiers in Neuroscience 7:181. 10.3389/fnins.2013.00181 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Söderholm JD, Yang P, Ceponis P, Vohra A, Riddell R, Sherman PM, Perdue MH (2002). Chronic stress induces mast cell-dependent bacterial adherence and initiates mucosal inflammation in rat intestine. Gastroenterology 123(4): 1099–1108. 10.1053/gast.2002.36019 [DOI] [PubMed] [Google Scholar]
- Stewart AS, Pratt-Phillips S, Gonzalez LM (2017). Alterations in Intestinal Permeability: The Role of the “Leaky Gut” in Health and Disease. Journal of Equine Veterinary Science 52:10–22. 10.1016/j.jevs.2017.02.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stilling RM, van de Wouw M, Clarke G, Stanton C, Dinan TG, Cryan JF (2016). The neuropharmacology of butyrate: the bread and butter of the microbiota-gut-brain axis? Neurochemistry International 99:110–132. [DOI] [PubMed] [Google Scholar]
- Sudo N (2019). Biogenic Amines: Signals Between Commensal Microbiota and Gut Physiology. Frontiers in Endocrinology 10:504. 10.3389/fendo.2019.00504 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu X, et al. (2004). Postnatal microbial colonization programs the hypothalamic–pituitary–adrenal system for stress response in mice. The Journal of Physiology 558(1):263–275. 10.1113/jphysiol.2004.063388 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun J, Xu J, Ling Y, Wang F, Gong T, Yang C, et al. (2019). Fecal microbiota transplantation alleviated Alzheimer’s disease-like pathogenesis in APP/PS1 transgenic mice. Translational Psychiatry 9(1): 189–189. 10.1038/s41398-019-0525-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan T, Kuramoto M, Takahashi T, Nakamura H, Nakanishi Y, Imasato Y, Yoshimura H (1989). Characteristics of the gastrointestinal absorption of morphine in rats. Chemical and Pharmaceutical Bulletin 37(1): 168–173. [DOI] [PubMed] [Google Scholar]
- Tarr AJ, Galley JD, Fisher SE, Chichlowski M, Berg BM, Bailey MT (2015). The prebiotics 3’Sialyllactose and 6’Sialyllactose diminish stressor-induced anxiety-like behavior and colonic microbiota alterations: Evidence for effects on the gut-brain axis. Brain, Behavior, and Immunity 50:166–177. 10.1016/j.bbi.2015.06.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Taylor AMW, Castonguay A, Ghogha A, Vayssiere P, Pradhan AAA, Xue L, et al. (2015). Neuroimmune Regulation of GABAergic Neurons Within the Ventral Tegmental Area During Withdrawal from Chronic Morphine. Neuropsychopharmacology 41:949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Temko JE, Bouhlal S, Farokhnia M, Lee MR, Cryan JF, Leggio L (2017). The Microbiota, the Gut and the Brain in Eating and Alcohol Use Disorders: A ‘Ménage à Trois’? Alcohol and Alcoholism 52(4):403–413. 10.1093/alcalc/agx024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Thomas CM, Hong T, van Pijkeren JP, Hemarajata P, Trinh DV, Hu W, et al. (2012). Histamine derived from probiotic Lactobacillus reuteri suppresses TNF via modulation of PKA and ERK signaling. PloS One 7(2):e31951–e31951. 10.1371/journal.pone.0031951 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tindell AJ, Smith KS, Berridge KC, Aldridge JW (2009). Dynamic computation of incentive salience: ‘wanting’ what was never ‘liked’. The Journal of Neuroscience 29(39): 12220–12228. 10.1523/JNEUROSCI.2499-09.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsankova N, Renthal W, Kumar A, Nestler EJ (2007). Epigenetic regulation in psychiatric disorders. Nature Reviews Neuroscience 8:355. [DOI] [PubMed] [Google Scholar]
- Tuesta LM, Chen Z, Duncan A, Fowler CD, Ishikawa M, Lee BR, et al. (2017). GLP-1 acts on habenular avoidance circuits to control nicotine intake. Nature Neuroscience 20(5):708–716. 10.1038/nn.4540 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vallöf D, Maccioni P, Colombo G, Mandrapa M, Jömulf JW, Egecioglu E, et al. (2016). The glucagon-like peptide 1 receptor agonist liraglutide attenuates the reinforcing properties of alcohol in rodents. Addiction Biology 21(2):422–437. 10.1111/adb.12295 [DOI] [PMC free article] [PubMed] [Google Scholar]
- van de Wouw M, Stilling RM, Peterson VL, Ryan FJ, Hoban AE, Shanahan F, et al. (2019). Host Microbiota Regulates Central Nervous System Serotonin Receptor 2C Editing in Rodents. ACS Chemical Neuroscience. 10.1021/acschemneuro.9b00414 [DOI] [PubMed] [Google Scholar]
- van der Laan LN, de Ridder DTD, Viergever MA, Smeets PAM (2014). Activation in inhibitory brain regions during food choice correlates with temptation strength and self-regulatory success in weight-concerned women. Frontiers in Neuroscience 8:308–308. 10.3389/fnins.2014.00308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vincent C, Miller MA, Edens TJ, Mehrotra S, Dewar K, Manges AR (2016). Bloom and bust: intestinal microbiota dynamics in response to hospital exposures and Clostridium difficile colonization or infection. Microbiome 4(1): 12. 10.1186/s40168-016-0156-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vindigni SM, Surawicz CM (2017). Fecal Microbiota Transplantation. The Gut Microbiome 46(1): 171–185. 10.1016/j.gtc.2016.09.012 [DOI] [PubMed] [Google Scholar]
- Volkow ND, Morales M (2015). The Brain on Drugs: From Reward to Addiction. Cell 162(4):712–725. 10.1016/j.cell.2015.07.046 [DOI] [PubMed] [Google Scholar]
- Volkow ND, Wise RA, Baler R (2017). The dopamine motive system: implications for drug and food addiction. Nature Reviews Neuroscience 18(12):741. [DOI] [PubMed] [Google Scholar]
- Volpe GE, Ward H, Mwamburi M, Dinh D, Bhalchandra S, Wanke C, Kane AV (2014). Associations of cocaine use and HIV infection with the intestinal microbiota, microbial translocation, and inflammation. Journal of Studies on Alcohol and Drugs 75(2):347–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wall R, Cryan JF, Ross RP, Fitzgerald GF, Dinan TG, Stanton C (2014). Bacterial Neuroactive Compounds Produced by Psychobiotics. In: Microbial Endocrinology: The Microbiota-Gut-Brain Axis in Elealth and Disease Lyte M, Cryan JF (editors), pp. 221–239. [DOI] [PubMed] [Google Scholar]
- Wang F, Meng J, Zhang L, Johnson T, Chen C, Roy S (2018). Morphine induces changes in the gut microbiome and metabolome in a morphine dependence model. Scientific Reports 8(1):3596. 10.1038/s41598-018-21915-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang F, Roy S (2016). Gut Homeostasis, Microbial Dysbiosis, and Opioids. Toxicologic Pathology 45(1): 150–156. 10.1177/0192623316679898 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wohleb ES, Delpech JC (2017). Dynamic cross-talk between microglia and peripheral monocytes underlies stress-induced neuroinflammation and behavioral consequences. Progress in Neuro-Psychopharmacology and Biological Psychiatry 79:40–48. [DOI] [PubMed] [Google Scholar]
- Wortelboer K, Nieuwdorp M, Herrema H (2019). Fecal microbiota transplantation beyond Clostridioides difficile infections. EBioMedicine 44:716–729. 10.1016/j.ebiom.2019.05.066 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xiao H, Ge C, Feng G, Li Y, Luo D, Dong J, et al. (2018). Gut microbiota modulates alcohol withdrawal-induced anxiety in mice. Toxicology Letters 287:23–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu Y, Xie Z, Wang H, Shen Z, Guo Y, Gao Y, et al. (2017). Bacterial diversity of intestinal microbiota in patients with substance use disorders revealed by 16S rRNA gene deep sequencing. Scientific Reports 7(1):3628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang C, Fang X, Zhan G, Huang N, Li S, Bi J, et al. (2019). Key role of gut microbiota in anhedonia-like phenotype in rodents with neuropathic pain. Translational Psychiatry 9(1):57. 10.1038/s41398-019-0379-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang L, Zhao Y, Wang Y, Liu L, Zhang X, Li B, Cui R (2015). The effects of psychological stress on depression. Current Neuropharmacology 13(4):494–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, et al. (2015). Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 161(2):264–276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yau YH, Potenza MN (2013). Stress and eating behaviors. Minerva Endocrinologica 38(3):255. [PMC free article] [PubMed] [Google Scholar]
- Zhang J, Berridge KC, Tindell AJ, Smith KS, Aldridge JW (2009). A Neural Computational Model of Incentive Salience. PLOS Computational Biology 5(7):el000437. 10.1371/journal.pcbi.1000437 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, et al. (2016). Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 352(6285):565. 10.1126/science.aad3369 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu F, Guo R, Wang W, Ju Y, Wang Q, Ma Q, et al. (2019). Transplantation of microbiota from drug-free patients with schizophrenia causes schizophrenia-like abnormal behaviors and dysregulated kynurenine metabolism in mice. Molecular Psychiatry, 10.1038/s41380-019-0475-4 [DOI] [PubMed] [Google Scholar]
- Zimmermann M, Zimmermann-Kogadeeva M, Wegmann R, Goodman AL (2019). Mapping human microbiome drug metabolism by gut bacteria and their genes. Nature 570(7762):462–467. 10.1038/s41586-019-1291-3 [DOI] [PMC free article] [PubMed] [Google Scholar]