1
|
Schneider E, Schmidt R, Cryan JF, Hilbert A. A Role for the Microbiota-Gut-Brain Axis in Avoidant/Restrictive Food Intake Disorder: A New Conceptual Model. Int J Eat Disord 2024. [PMID: 39542726 DOI: 10.1002/eat.24326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE Avoidant/restrictive food intake disorder (ARFID) is an eating disorder characterized by a severely restrictive diet leading to significant physical and/or psychosocial sequelae. Largely owing to the phenotypic heterogeneity, the underlying pathophysiological mechanisms are relatively unknown. Recently, the communication between microorganisms within the gastrointestinal tract and the brain-the so-called microbiota-gut-brain axis-has been implicated in the pathophysiology of eating disorders. This Spotlight review sought to investigate and conceptualize the possible ways that the microbiota-gut-brain axis is involved in ARFID to drive future research in this area. METHOD By relating core symptoms of ARFID to gut microbiota and its signaling pathways to the brain, we evaluated how the gut microbiota is potentially involved in the pathophysiology of ARFID. RESULTS We hypothesized that the restricted type and amount of food intake characteristic of ARFID diminishes gut microbial diversity, including beneficial bacteria and their metabolites capable of signaling to the brain, to modulate biopsychological pathways relevant to ARFID: homeostatic signaling, food reward, interoception, sensory sensitivity, disgust, perseveration, fear-based learning, and mood. Candidate signaling mechanisms include microbial-induced effects on inflammation, cortisol, and neurotransmitters such as dopamine and serotonin. DISCUSSION Through reviewing the extant evidence, we conceptualized a new theoretical framework of ARFID with an emphasis on microbiota-gut-brain axis signaling to inform future research. Although more research is necessary to evaluate this theoretical model, the tentative evidence suggests that therapeutics specifically targeting the gut microbiota for the treatment of ARFID symptomatology warrants more investigation.
Collapse
Affiliation(s)
| | - Ricarda Schmidt
- Department of Psychosomatic Medicine and Psychotherapy, Research Unit Behavioral Medicine, Integrated Research and Treatment Center AdiposityDiseases, University of Leipzig Medical Center, Leipzig, Germany
- German Center for Child and Adolescent Health (DZKJ), partner Site Leipzig/Dresden, Leipzig, Germany
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Anja Hilbert
- Department of Psychosomatic Medicine and Psychotherapy, Research Unit Behavioral Medicine, Integrated Research and Treatment Center AdiposityDiseases, University of Leipzig Medical Center, Leipzig, Germany
- German Center for Child and Adolescent Health (DZKJ), partner Site Leipzig/Dresden, Leipzig, Germany
| |
Collapse
|
2
|
Karaivazoglou K, Aggeletopoulou I, Triantos C. The Contribution of the Brain-Gut Axis to the Human Reward System. Biomedicines 2024; 12:1861. [PMID: 39200325 PMCID: PMC11351993 DOI: 10.3390/biomedicines12081861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
The human reward network consists of interconnected brain regions that process stimuli associated with satisfaction and modulate pleasure-seeking behaviors. Impairments in reward processing have been implicated in several medical and psychiatric conditions, and there is a growing interest in disentangling the underlying pathophysiological mechanisms. The brain-gut axis plays a regulatory role in several higher-order neurophysiological pathways, including reward processing. In this context, the aim of the current review was to critically appraise research findings on the contribution of the brain-gut axis to the human reward system. Enteric neuropeptides, which are implicated in the regulation of hunger and satiety, such as ghrelin, PYY3-36, and glucagon-like peptide 1 (GLP-1), have been associated with the processing of food-related, alcohol-related, and other non-food-related rewards, maintaining a delicate balance between the body's homeostatic and hedonic needs. Furthermore, intestinal microbiota and their metabolites have been linked to differences in the architecture and activation of brain reward areas in obese patients and patients with attention deficit and hyperactivity disorder. Likewise, bariatric surgery reduces hedonic eating by altering the composition of gut microbiota. Although existing findings need further corroboration, they provide valuable information on the pathophysiology of reward-processing impairments and delineate a novel framework for potential therapeutic interventions.
Collapse
Affiliation(s)
| | - Ioanna Aggeletopoulou
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece;
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece;
| |
Collapse
|
3
|
Schneider E, O'Riordan KJ, Clarke G, Cryan JF. Feeding gut microbes to nourish the brain: unravelling the diet-microbiota-gut-brain axis. Nat Metab 2024; 6:1454-1478. [PMID: 39174768 DOI: 10.1038/s42255-024-01108-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 07/15/2024] [Indexed: 08/24/2024]
Abstract
The prevalence of brain disorders, including stress-related neuropsychiatric disorders and conditions with cognitive dysfunction, is rising. Poor dietary habits contribute substantially to this accelerating trend. Conversely, healthy dietary intake supports mood and cognitive performance. Recently, the communication between the microorganisms within the gastrointestinal tract and the brain along the gut-brain axis has gained prominence as a potential tractable target to modulate brain health. The composition and function of the gut microbiota is robustly influenced by dietary factors to alter gut-brain signalling. To reflect this interconnection between diet, gut microbiota and brain functioning, we propose that a diet-microbiota-gut-brain axis exists that underpins health and well-being. In this Review, we provide a comprehensive overview of the interplay between diet and gut microbiota composition and function and the implications for cognition and emotional functioning. Important diet-induced effects on the gut microbiota for the development, prevention and maintenance of neuropsychiatric disorders are described. The diet-microbiota-gut-brain axis represents an uncharted frontier for brain health diagnostics and therapeutics across the lifespan.
Collapse
Affiliation(s)
| | | | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
4
|
Lee B, Lee SM, Song JW, Choi JW. Gut Microbiota Metabolite Messengers in Brain Function and Pathology at a View of Cell Type-Based Receptor and Enzyme Reaction. Biomol Ther (Seoul) 2024; 32:403-423. [PMID: 38898687 PMCID: PMC11214962 DOI: 10.4062/biomolther.2024.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/02/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
The human gastrointestinal (GI) tract houses a diverse microbial community, known as the gut microbiome comprising bacteria, viruses, fungi, and protozoa. The gut microbiome plays a crucial role in maintaining the body's equilibrium and has recently been discovered to influence the functioning of the central nervous system (CNS). The communication between the nervous system and the GI tract occurs through a two-way network called the gut-brain axis. The nervous system and the GI tract can modulate each other through activated neuronal cells, the immune system, and metabolites produced by the gut microbiome. Extensive research both in preclinical and clinical realms, has highlighted the complex relationship between the gut and diseases associated with the CNS, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. This review aims to delineate receptor and target enzymes linked with gut microbiota metabolites and explore their specific roles within the brain, particularly their impact on CNS-related diseases.
Collapse
Affiliation(s)
- Bada Lee
- Department of Biomedicinal and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Soo Min Lee
- Department of Biomedicinal and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae Won Song
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jin Woo Choi
- Department of Biomedicinal and Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Regulatory Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
5
|
Hameed M, Noor F, Hussain H, Khan RG, Khattak Haroon Ur Rashid S, Haroon Ur Rashid S, Atiq A, Ali H, Rida SE, Abbasi MA. Gut-Brain Axis: Investigating the Effects of Gut Health on Cognitive Functioning in Adults. Cureus 2024; 16:e64286. [PMID: 39130956 PMCID: PMC11315957 DOI: 10.7759/cureus.64286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
INTRODUCTION The gut-brain axis is a bidirectional communication network linking the gastrointestinal tract and the central nervous system via neuronal, hormonal, and antibody signaling pathways. Central to this connection is gut health, encompassing the balance and functionality of gut microbiota, which significantly impacts on mental and cognitive health. This study investigates the association between gut health and cognitive functioning in adults, highlighting the mechanisms by which gut microbiota influence brain health. OBJECTIVE To examine the effects of gut health on adult cognitive performance, with a focus on the processes by which gut microbiota impacts brain health. METHODS A quantitative cross-sectional study was conducted in Islamabad from January 2024 to April 2024, involving 140 adult participants. Data were collected using a comprehensive 16-item gut health questionnaire and the cognition self-assessment rating scale (C-SARS). The psychometric properties of these scales were assessed, and the data were analyzed using Statistical Product and Service Solutions (SPSS, v26; IBM SPSS Statistics for Windows, Armonk, NY). Analytical and descriptive statistics, including regression, chi-square, independent sample t-tests, and mean and standard deviation, were applied. RESULTS The study found moderate associations between gut health and cognitive performance, particularly in memory and processing speed (R² = 0.17, β = -1.9, p = 0.12 for general cognition; R² = 0.01, β = -0.98, p = 0.02 for memory; R² = 0.03, β = -0.18, p = 0.03 for processing speed). Gender and marital status differences were significant, with males exhibiting better gut health scores than females (M = 34.1, SD = 3.2 vs. M = 31.2, SD = 3.2, p = 0.00), and singles showing better cognitive performance compared to married individuals (M = 9.4, SD = 5.4 vs. M = 6.5, SD = 3.7, p = 0.03). CONCLUSION The study highlights significant associations between gut health and cognitive functions, suggesting that gut microbiota composition can influence cognitive performance. Gender and marital status differences underscore the need to consider individual differences in gut-brain axis research. Future studies should replicate these findings in larger samples and explore gut microbiota-targeted interventions for cognitive health enhancement.
Collapse
Affiliation(s)
- Muddsar Hameed
- Department of Clinical Psychology, Shifa Tameer-e-Millat University, Islamabad, PAK
| | - Fatima Noor
- Department of Internal Medicine, Foundation University Medical College, Islamabad, PAK
| | - Hamza Hussain
- Department of Internal Medicine, Foundation University Medical College, Islamabad, PAK
| | - Raja Gohar Khan
- Department of Internal Medicine, Foundation University Medical College, Islamabad, PAK
| | | | | | - Alina Atiq
- Department of Internal Medicine, Al Nafees Medical College and Hospital, Islamabad, PAK
| | - Hassan Ali
- Department of Psychology, Birmingham City University, Birmingham, GBR
| | - Seerat E Rida
- Department of Internal Medicine, Bahria University Medical and Dental College, Karachi, PAK
| | - Mahrukh Anwar Abbasi
- Department of Internal Medicine, Foundation University Medical College, Islamabad, PAK
| |
Collapse
|
6
|
Wei B, Peng Z, Zheng W, Yang S, Wu M, Liu K, Xiao M, Huang T, Xie M, Xiong T. Probiotic-fermented tomato alleviates high-fat diet-induced obesity in mice: Insights from microbiome and metabolomics. Food Chem 2024; 436:137719. [PMID: 37839120 DOI: 10.1016/j.foodchem.2023.137719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/02/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023]
Abstract
Probiotic-fermented plant-based foods are associated with weight loss. Here, we hypothesized probiotic-fermented tomato (FT) as a functional food with potential to alleviate obesity, thus the obesity-alleviating effects and mechanisms of FT on high-fat diet-induced obese mice were explored via biochemical, gut microbiome, and serum metabolomics analysis. The results showed that FT performed better than unfermented tomato in reducing body weight gain and fat accumulation, improving dyslipidemia and glucose homeostasis, and relieving inflammation and adipocytokine dysregulation. Particularly, live probiotic-fermented tomato (LFT) was associated with improved diversity, composition, and structure of gut microbiota, suppressed obesity-related genera growth (e.g., Clostridium, Olsenella, and Mucispirillum), and promoted beneficial genera growth (e.g., Roseburia, Coprococcus, and Oscillospira), which were associated negatively with body weight, TC, TG, and TNF-α levels. Additionally, LFT was associated with positive changes in glycerophospholipids, sphingolipids, unsaturated fatty acids, and amino acids levels. Collectively, as a functional food, LFT possessed potential for obesity alleviation.
Collapse
Affiliation(s)
- Benliang Wei
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Zhen Peng
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Wendi Zheng
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Shiyu Yang
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Min Wu
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Kui Liu
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Muyan Xiao
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; International Institute of Food Innovation, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Tao Huang
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; International Institute of Food Innovation, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Mingyong Xie
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Tao Xiong
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China.
| |
Collapse
|
7
|
Speckmann B, Ehring E, Hu J, Rodriguez Mateos A. Exploring substrate-microbe interactions: a metabiotic approach toward developing targeted synbiotic compositions. Gut Microbes 2024; 16:2305716. [PMID: 38300741 PMCID: PMC10841028 DOI: 10.1080/19490976.2024.2305716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
Gut microbiota is an important modulator of human health and contributes to high inter-individual variation in response to food and pharmaceutical ingredients. The clinical outcomes of interventions with prebiotics, probiotics, and synbiotics have been mixed and often unpredictable, arguing for novel approaches for developing microbiome-targeted therapeutics. Here, we review how the gut microbiota determines the fate of and individual responses to dietary and xenobiotic compounds via its immense metabolic potential. We highlight that microbial metabolites play a crucial role as targetable mediators in the microbiota-host health relationship. With this in mind, we expand the concept of synbiotics beyond prebiotics' role in facilitating growth and engraftment of probiotics, by focusing on microbial metabolism as a vital mode of action thereof. Consequently, we discuss synbiotic compositions that enable the guided metabolism of dietary or co-formulated ingredients by specific microbes leading to target molecules with beneficial functions. A workflow to develop novel synbiotics is presented, including the selection of promising target metabolites (e.g. equol, urolithin A, spermidine, indole-3 derivatives), identification of suitable substrates and producer strains applying bioinformatic tools, gut models, and eventually human trials.In conclusion, we propose that discovering and enabling specific substrate-microbe interactions is a valuable strategy to rationally design synbiotics that could establish a new category of hybrid nutra-/pharmaceuticals.
Collapse
Affiliation(s)
| | | | - Jiaying Hu
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Ana Rodriguez Mateos
- Department of Nutritional Sciences, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, UK
| |
Collapse
|
8
|
Purdel C, Margină D, Adam-Dima I, Ungurianu A. The Beneficial Effects of Dietary Interventions on Gut Microbiota-An Up-to-Date Critical Review and Future Perspectives. Nutrients 2023; 15:5005. [PMID: 38068863 PMCID: PMC10708505 DOI: 10.3390/nu15235005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/22/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Different dietary interventions, especially intermittent fasting, are widely used and promoted by physicians; these regimens have been studied lately for their impact on the gut microbiota composition/function and, consequently, on the general physiopathological processes of the host. Studies are showing that dietary components modulate the microbiota, and, at the same time, the host metabolism is deeply influenced by the different products resulting from nutrient transformation in the microbiota compartment. This reciprocal relationship can potentially influence even drug metabolism for chronic drug regimens, significantly impacting human health/disease. Recently, the influence of various dietary restrictions on the gut microbiota and the differences between the effects were investigated. In this review, we explored the current knowledge of different dietary restrictions on animal and human gut microbiota and the impact of these changes on human health.
Collapse
Affiliation(s)
- Carmen Purdel
- Department of Toxicology, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (C.P.); (I.A.-D.)
| | - Denisa Margină
- Department of Biochemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania;
| | - Ines Adam-Dima
- Department of Toxicology, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (C.P.); (I.A.-D.)
| | - Anca Ungurianu
- Department of Biochemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania;
| |
Collapse
|
9
|
Wang J, Zhou T, Liu F, Huang Y, Xiao Z, Qian Y, Zhou W. Influence of gut microbiota on resilience and its possible mechanisms. Int J Biol Sci 2023; 19:2588-2598. [PMID: 37215996 PMCID: PMC10197883 DOI: 10.7150/ijbs.82362] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/28/2023] [Indexed: 05/24/2023] Open
Abstract
Excessive stress leads to disruptions of the central nervous system. Individuals' responses to stress and trauma differ from person to person. Some may develop various neuropsychiatric disorders, such as post-traumatic stress disorder, major depression, and anxiety disorders, while others may successfully adapt to the same stressful events. These two neural phenotypes are called susceptibility and resilience. Previous studies have suggested resilience/susceptibility as a complex, non-specific systemic response involving central and peripheral systems. Emerging research of mechanisms underlying resilience is mostly focussing on the physiological adaptation of specific brain circuits, neurovascular impairment of the blood-brain barrier, the role of innate and adaptive factors of the immune system, and the dysbiosis of gut microbiota. In accordance with the microbiota-gut-brain axis hypothesis, the gut microbiome directly influences the interface between the brain and the periphery to affect neuronal function. This review explored several up-to-date studies on the role of gut microbiota implicated in stressful events-related resilience/susceptibility. We mainly focus on the changes in behavior and neuroimaging characteristics, involved brain regions and circuits, the blood-brain barrier, the immune system, and epigenetic modifications, which contribute to stress-induced resilience and susceptibility. The perspective of the gut-brain axis could help to understand the mechanisms underlying resilience and the discovery of biomarkers may lead to new research directions and therapeutic interventions for stress-induced neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jianhui Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Ting Zhou
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Feng Liu
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Yan Huang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Zhiyong Xiao
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| | - Yan Qian
- Department of Pharmacy, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Wenxia Zhou
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China
| |
Collapse
|
10
|
Osadchiy V, Bal R, Mayer EA, Kunapuli R, Dong T, Vora P, Petrasek D, Liu C, Stains J, Gupta A. Machine learning model to predict obesity using gut metabolite and brain microstructure data. Sci Rep 2023; 13:5488. [PMID: 37016129 PMCID: PMC10073225 DOI: 10.1038/s41598-023-32713-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 03/31/2023] [Indexed: 04/06/2023] Open
Abstract
A growing body of preclinical and clinical literature suggests that brain-gut-microbiota interactions may contribute to obesity pathogenesis. In this study, we use a machine learning approach to leverage the enormous amount of microstructural neuroimaging and fecal metabolomic data to better understand key drivers of the obese compared to overweight phenotype. Our findings reveal that although gut-derived factors play a role in this distinction, it is primarily brain-directed changes that differentiate obese from overweight individuals. Of the key gut metabolites that emerged from our model, many are likely at least in part derived or influenced by the gut-microbiota, including some amino-acid derivatives. Remarkably, key regions outside of the central nervous system extended reward network emerged as important differentiators, suggesting a role for previously unexplored neural pathways in the pathogenesis of obesity.
Collapse
Affiliation(s)
- Vadim Osadchiy
- Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, USA
- UCLA Microbiome Center, Los Angeles, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Los Angeles, USA
- Department of Urology, David Geffen School of Medicine, Los Angeles, USA
| | - Roshan Bal
- Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, USA
| | - Emeran A Mayer
- Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, USA
- UCLA Microbiome Center, Los Angeles, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Los Angeles, USA
| | - Rama Kunapuli
- Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, USA
| | - Tien Dong
- Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, USA
- UCLA Microbiome Center, Los Angeles, USA
| | - Priten Vora
- Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Los Angeles, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Danny Petrasek
- Department of Mathematics, California Institute of Technology, Pasadena, USA
| | - Cathy Liu
- Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Los Angeles, USA
| | - Jean Stains
- Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, USA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Los Angeles, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Arpana Gupta
- Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, USA.
- UCLA Microbiome Center, Los Angeles, USA.
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Los Angeles, USA.
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, CHS 42-210 MC737818, 10833 Le Conte Avenue, Los Angeles, CA, USA.
| |
Collapse
|
11
|
Sarnoff RP, Bhatt RR, Osadchiy V, Dong T, Labus JS, Kilpatrick LA, Chen Z, Subramanyam V, Zhang Y, Ellingson BM, Naliboff B, Chang L, Mayer EA, Gupta A. A multi-omic brain gut microbiome signature differs between IBS subjects with different bowel habits. Neuropharmacology 2023; 225:109381. [PMID: 36539012 DOI: 10.1016/j.neuropharm.2022.109381] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/25/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Alterations of the brain-gut-microbiome system (BGM) have been implicated in the pathophysiology of irritable bowel syndrome (IBS), yet bowel habit-specific alterations have not been elucidated. In this cross-sectional study, we apply a systems biology approach to characterize BGM patterns related to predominant bowel habit. Fecal samples and resting state fMRI were obtained from 102 premenopausal women (36 constipation-predominant IBS (IBS-C), 27 diarrhea-predominant IBS (IBS-D), 39 healthy controls (HCs)). Data integration analysis using latent components (DIABLO) was used to integrate data from the phenome, microbiome, metabolome, and resting-state connectome to predict HCs vs IBS-C vs IBS-D. Bloating and visceral sensitivity, distinguishing IBS from HC, were negatively associated with beneficial microbes and connectivity involving the orbitofrontal cortex. This suggests that gut interactions may generate aberrant central autonomic and descending pain pathways in IBS. The connection between IBS symptom duration, key microbes, and caudate connectivity may provide mechanistic insight to the chronicity of pain in IBS. Compared to IBS-C and HCs, IBS-D had higher levels of many key metabolites including tryptophan and phenylalanine, and increased connectivity between the sensorimotor and default mode networks; thus, suggestingan influence on diarrhea, self-related thoughts, and pain perception in IBS-D ('bottom-up' mechanism). IBS-C's microbiome and metabolome resembled HCs, but IBS-C had increased connectivity in the default mode and salience networks compared to IBS-D, which may indicate importance of visceral signals, suggesting a more 'top-down' BGM pathophysiology. These BGM characteristics highlight possible mechanistic differences for variations in the IBS bowel habit phenome. This article is part of the Special Issue on 'Microbiome & the Brain: Mechanisms & Maladies'.
Collapse
Affiliation(s)
- Rachel P Sarnoff
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Ravi R Bhatt
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, USA
| | - Vadim Osadchiy
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Tien Dong
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA; UCLA Microbiome Center, USA; Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Jennifer S Labus
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA
| | - Lisa A Kilpatrick
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA
| | - Zixi Chen
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA
| | | | - Yurui Zhang
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA
| | - Benjamin M Ellingson
- Departments of Radiological Sciences, Psychiatry, and Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Bruce Naliboff
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA
| | - Lin Chang
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA
| | - Emeran A Mayer
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA; UCLA Microbiome Center, USA.
| | - Arpana Gupta
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, USA; David Geffen School of Medicine, USA; Vatche and Tamar Manoukian Division of Digestive Diseases, USA; UCLA Microbiome Center, USA.
| |
Collapse
|
12
|
Yang H, Yang K, Zhang L, Yang N, Mei YX, Zheng YL, He Y, Gong YJ, Ding WJ. Acupuncture ameliorates Mobile Phone Addiction with sleep disorders and restores salivary metabolites rhythm. Front Psychiatry 2023; 14:1106100. [PMID: 36896350 PMCID: PMC9989025 DOI: 10.3389/fpsyt.2023.1106100] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/03/2023] [Indexed: 02/25/2023] Open
Abstract
OBJECTIVES Mobile Phone Addiction (MPA) is a novel behavioral addiction resulting in circadian rhythm disorders that severely affect mental and physical health. The purpose of this study is to detect rhythmic salivary metabolites in MPA with sleep disorder (MPASD) subjects and investigate the effects of acupuncture. METHODS Six MPASD patients and six healthy controls among the volunteers were enrolled by MPA Tendency Scale (MPATS) and Pittsburgh Sleep Quality Index (PSQI), then the salivary samples of MPASD and healthy controls were collected every 4-h for three consecutive days. Acupuncture was administered for 7 days to MPASD subjects, then saliva samples were collected again. Salivary metabolomes were analyzed with the method of LC-MS. RESULT According to our investigation, 70 (57.85%) MPA patients and 56 (46.28%) MPASD patients were identified among 121 volunteers. The symptoms of the 6 MPASD subjects were significantly alleviated after acupuncture intervention. The number of rhythmic saliva metabolites dropped sharply in MPASD subjects and restored after acupuncture. Representative rhythmic saliva metabolites including melatonin, 2'-deoxyuridine, thymidine, thymidine 3',5'-cyclic monophosphate lost rhythm and restored after acupuncture, which may attribute to promising MPASD treatment and diagnosis biomarkers. The rhythmic saliva metabolites of healthy controls were mainly enriched in neuroactive ligand-receptor interaction, whereas polyketide sugar unit biosynthesis was mainly enriched in MPASD patients. CONCLUSION This study revealed circadian rhythm characteristics of salivary metabolites in MPASD and that acupuncture could ameliorate MPASD by restoring part of the dysrhythmia salivary metabolites.
Collapse
Affiliation(s)
- Hong Yang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kun Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Zhang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ni Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying-Xiu Mei
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ya-Li Zheng
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan He
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan-Ju Gong
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei-Jun Ding
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
13
|
Xavier J, Anu M, Fathima AS, Ravichandiran V, Kumar N. Intriguing Role of Gut-Brain Axis on Cognition with an Emphasis on Interaction with Papez Circuit. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:1146-1163. [PMID: 35702801 DOI: 10.2174/1871527321666220614124145] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 06/15/2023]
Abstract
The gut microbiome is a complicated ecosystem of around a hundred billion symbiotic bacteria cells. Bidirectional communication between the gut and the brain is facilitated by the immune system, the enteric nervous system, the vagus nerve, and microbial compounds such as tryptophan metabolites and short-chain fatty acids (SCFAs). The current study emphasises the relationship of the gut-brain axis with cognitive performance and elucidates the underlying biological components, with a focus on neurotransmitters such as serotonin, indole derivatives, and catecholamine. These biological components play important roles in both the digestive and brain systems. Recent research has linked the gut microbiome to a variety of cognitive disorders, including Alzheimer's (AD). The review describes the intriguing role of the gut-brain axis in recognition memory depending on local network connections within the hippocampal as well as other additional hippocampal portions of the Papez circuit. The available data from various research papers show how the gut microbiota might alter brain function and hence psychotic and cognitive illnesses. The role of supplementary probiotics is emphasized for the reduction of brain-related dysfunction as a viable strategy in handling cognitive disorders. Further, the study elucidates the mode of action of probiotics with reported adverse effects.
Collapse
Affiliation(s)
- Joyal Xavier
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, India
| | - M Anu
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur, India
| | - A S Fathima
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Hajipur, Vaishali- 844102, Bihar, India
| | - V Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur, India
| |
Collapse
|
14
|
Lai Y, Dhingra R, Zhang Z, Ball LM, Zylka MJ, Lu K. Toward Elucidating the Human Gut Microbiota-Brain Axis: Molecules, Biochemistry, and Implications for Health and Diseases. Biochemistry 2022; 61:2806-2821. [PMID: 34910469 PMCID: PMC10857864 DOI: 10.1021/acs.biochem.1c00656] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent years, a substantial amount of data have supported an active role of gut microbiota in mediating mammalian brain function and health. Mining gut microbiota and their metabolites for neuroprotection is enticing but requires that the fundamental biochemical details underlying such microbiota-brain crosstalk be deciphered. While a neuronal gut-brain axis (through the vagus nerve) is not disputable, accumulating studies also point to a humoral route (via blood/lymphatic circulation) by which innumerable microbial molecular cues translocate from local gut epithelia to circulation with potentials to further cross the blood-brain barrier and reach the brain. In this Perspective, we review a realm of gut microbial molecules to evaluate their fate, function, and neuroactivities in vivo as mediated by microbiota. We turn to seminal studies of neurophysiology and neurologic disease models for the elucidation of biochemical pathways that link microbiota to gut-brain signaling. In addition, we discuss opportunities and challenges for advancing the microbiota-brain axis field while calling for high-throughput discovery of microbial molecules and studies for resolving the interspecies, interorgan, and interclass interaction among these neuroactive microbial molecules.
Collapse
Affiliation(s)
- Yunjia Lai
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Campus Box 7431, Chapel Hill, North Carolina 27599, United States
| | - Radhika Dhingra
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Campus Box 7431, Chapel Hill, North Carolina 27599, United States
- Institute of Environmental Health Solutions, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Zhenfa Zhang
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Campus Box 7431, Chapel Hill, North Carolina 27599, United States
| | - Louise M Ball
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Campus Box 7431, Chapel Hill, North Carolina 27599, United States
| | - Mark J Zylka
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Carolina Institute for Developmental Disabilities, The University of North Carolina at Chapel Hill, Carrboro, North Carolina 27510, United States
- Department of Cell and Molecular Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Kun Lu
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Campus Box 7431, Chapel Hill, North Carolina 27599, United States
- Curriculum in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
15
|
Examining the Influence of the Human Gut Microbiota on Cognition and Stress: A Systematic Review of the Literature. Nutrients 2022; 14:nu14214623. [PMID: 36364881 PMCID: PMC9656545 DOI: 10.3390/nu14214623] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
The gut microbiota is seen as an emerging biotechnology that can be manipulated to enhance or preserve cognition and physiological outputs of anxiety and depression in clinical conditions. However, the existence of such interactions in healthy young individuals in both non-stressful and stressful environments is unclear. The aim of this systematic review was to examine the relationship between the human gut microbiota, including modulators of the microbiota on cognition, brain function and/or stress, anxiety and depression. A total of n = 25 eligible research articles from a possible 3853 published between October 2018 and August 2021 were identified and included. Two study design methods for synthesis were identified: cross-sectional or pre/post intervention. Few cross-sectional design studies that linked microbiota to cognition, brain activity/structure or mental wellbeing endpoints existed (n = 6); however, correlations between microbiota diversity and composition and areas of the brain related to cognitive functions (memory and visual processing) were observed. Intervention studies targeting the gut microbiota to improve cognition, brain structure/function or emotional well-being (n = 19) generally resulted in improved brain activity and/or cognition (6/8), and improvements in depression and anxiety scores (5/8). Despite inherit limitations in studies reviewed, available evidence suggests that gut microbiota is linked to brain connectivity and cognitive performance and that modulation of gut microbiota could be a promising strategy for enhancing cognition and emotional well-being in stressed and non-stressed situations.
Collapse
|
16
|
Guerrero-Hreins E, Foldi CJ, Oldfield BJ, Stefanidis A, Sumithran P, Brown RM. Gut-brain mechanisms underlying changes in disordered eating behaviour after bariatric surgery: a review. Rev Endocr Metab Disord 2022; 23:733-751. [PMID: 34851508 DOI: 10.1007/s11154-021-09696-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/12/2021] [Indexed: 02/07/2023]
Abstract
Bariatric surgery results in long-term weight loss and an improved metabolic phenotype due to changes in the gut-brain axis regulating appetite and glycaemia. Neuroendocrine alterations associated with bariatric surgery may also influence hedonic aspects of eating by inducing changes in taste preferences and central reward reactivity towards palatable food. However, the impact of bariatric surgery on disordered eating behaviours (e.g.: binge eating, loss-of-control eating, emotional eating and 'addictive eating'), which are commonly present in people with obesity are not well understood. Increasing evidence suggests gut-derived signals, such as appetitive hormones, bile acid profiles, microbiota concentrations and associated neuromodulatory metabolites, can influence pathways in the brain implicated in food intake, including brain areas involved in sensorimotor, reward-motivational, emotional-arousal and executive control components of food intake. As disordered eating prevalence is a key mediator of weight-loss success and patient well-being after bariatric surgery, understanding how changes in the gut-brain axis contribute to disordered eating incidence and severity after bariatric surgery is crucial to better improve treatment outcomes in people with obesity.
Collapse
Affiliation(s)
- Eva Guerrero-Hreins
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Melbourne, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia
| | - Claire J Foldi
- Department of Physiology, Monash University, Clayton, Melbourne, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Australia
| | - Brian J Oldfield
- Department of Physiology, Monash University, Clayton, Melbourne, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Australia
| | - Aneta Stefanidis
- Department of Physiology, Monash University, Clayton, Melbourne, Australia
- Biomedicine Discovery Institute, Monash University, Clayton, Melbourne, Australia
| | - Priya Sumithran
- Department of Medicine (St Vincent's), University of Melbourne, Melbourne, Australia
- Department of Endocrinology, Austin Health, Melbourne, Australia
| | - Robyn M Brown
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Melbourne, Australia.
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, Australia.
| |
Collapse
|
17
|
Microbiome-metabolomics insights into the feces of high-fat diet mice to reveal the anti-obesity effects of yak (Bos grunniens) bone collagen hydrolysates. Food Res Int 2022; 156:111024. [DOI: 10.1016/j.foodres.2022.111024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/22/2022] [Accepted: 02/14/2022] [Indexed: 12/14/2022]
|
18
|
Perspective: Gestational Tryptophan Fluctuation Altering Neuroembryogenesis and Psychosocial Development. Cells 2022; 11:cells11081270. [PMID: 35455949 PMCID: PMC9032700 DOI: 10.3390/cells11081270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/10/2022] Open
Abstract
Tryptophan, as the sole precursor of serotonin, mainly derived from diets, is essential for neurodevelopment and immunomodulation. Gestational tryptophan fluctuation may account for the maternal-fetal transmission in determining neuroembryogenesis with long-lasting effects on psychological development. Personality disorders and social exclusion are related to psychosocial problems, leading to impaired social functioning. However, it is not clear how the fluctuation in mother-child transmission regulates the neuroendocrine development and gut microbiota composition in progeny due to that tryptophan metabolism in pregnant women is affected by multiple factors, such as diets (tryptophan-enriched or -depleted diet), emotional mental states (anxiety, depression), health status (hypertension, diabetes), and social support as well as stresses and management skills. Recently, we have developed a non-mammal model to rationalize those discrepancies without maternal effects. This perspective article outlines the possibility and verified the hypothesis in bully-victim research with this novel model: (1). Summarizes the effects of the maternal tryptophan administration on the neuroendocrine and microbial development in their offspring; (2). Highlights the inconsistency and limitations in studying the relationship between gestational tryptophan exposure and psychosocial development in humans and viviparous animals; and (3). Evidences that embryonic exposure to tryptophan and its metabolite modify bullying interactions in the chicken model. With the current pioneer researches on the biomechanisms underlying the bully-victim interaction, the perspective article provides novel insights for developing appropriate intervention strategies to prevent psychological disorders among individuals, especially those who experienced prenatal stress, by controlling dietary tryptophan and medication therapy during pregnancy.
Collapse
|
19
|
Huang X, Hu J, Peng H, Cheng HW. Embryonic Exposure to Tryptophan Yields Bullying Victimization via Reprogramming the Microbiota-Gut-Brain Axis in a Chicken Model. Nutrients 2022; 14:nu14030661. [PMID: 35277020 PMCID: PMC8839409 DOI: 10.3390/nu14030661] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/26/2022] [Accepted: 01/30/2022] [Indexed: 02/04/2023] Open
Abstract
Maternal metabolic disorder during early pregnancy may give rise to emotional and behavioral disorders in the child, vulnerable to bullying. Placental tryptophan fluctuation consequently disrupts offspring gut microbiome and brain neurogenesis with long-lasting physiological and social behavioral impacts. The aim of this study was to examine the hypothesis that the excess gestational tryptophan may affect children’s mental and physical development via modifying the microbiota-gut-brain axis, which lays the foundation of their mental status. Chicken embryo was employed due to its robust microbiota and independence of maternal influences during embryogenesis. The results indicated that embryonic tryptophan exposure reduced body weight and aggressiveness in the male offspring before and during adolescence. Additionally, the relative gut length and crypt depth were increased, while the villus/crypt ratio was decreased in tryptophan treated roosters, which was corresponding to the changes in the cecal microbiota composition. Furthermore, the catecholamine concentrations were increased in tryptophan group, which may be associated with the alterations in the gut microbiome and the gut-brain axis’s function. These changes may underlie the sociometric status of bullying; clarify how gestational tryptophan fluctuation compromises bullying and provide a strategy to prevent bullying by controlling dietary tryptophan and medication therapy during pregnancy.
Collapse
Affiliation(s)
- Xiaohong Huang
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao 266071, China
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.H.); (H.-w.C.)
- Correspondence: ; Tel.: +86-15908942478
| | - Jiaying Hu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.H.); (H.-w.C.)
| | - Haining Peng
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266071, China;
| | - Heng-wei Cheng
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.H.); (H.-w.C.)
- Livestock Behavior Research Unit, USDA-ARS, West Lafayette, IN 47907, USA
| |
Collapse
|
20
|
Liu D, Wang J, Zeng H, Zhou F, Wen B, Zhang X, Luo Y, Wu W, Huang J, Liu Z. The metabolic regulation of Fuzhuan brick tea in high-fat diet-induced obese mice and the potential contribution of gut microbiota. Food Funct 2022; 13:356-374. [PMID: 34904994 DOI: 10.1039/d1fo02181h] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This study investigated the metabolic effects of Fuzhuan brick tea (FBT) in high-fat diet (HFD)-induced obese mice and the potential contribution of gut microbiota. The results showed that FBT ameliorated the HFD-induced glycerophospholipid metabolic aberrance, specifically increased the serum levels of phosphatidylcholines (PCs), lysophosphatidylcholines (LysoPCs), and the ratio of PC to phosphatidylethanolamines (PE). Besides, FBT increased the serum level of gut microbiota-derived aryl hydrocarbon receptor (AhR) ligand, 3-indole propionic acid, as well as the relative abundance of intestinal AhR-ligand producing bacteria such as Clostridiaceae, Bacteroidales_S24-7_group, and Lactobacillaceae. However, the metabolic benefits of FBT were weakened when the gut microbiota were depleted by antibiotic treatment, thereby suggesting that gut microbiota was required for FBT to regulate glycerophospholipid metabolism. Indeed, the metabolites regulated by FBT were significantly correlated with the AhR-ligand producing bacteria. The KEGG pathway enrichment analysis and expressions of AhR target genes indicated that FBT would improve the glycerophospholipid metabolism via the AhR-Pemt signal axis, in which the gut microbiota and their metabolites played pivotal mediators. Overall, FBT could be a functional beverage to improve HFD-induced metabolic disorders in a gut microbiota dependent manner.
Collapse
Affiliation(s)
- Dongmin Liu
- Changsha University of Science & Technology, Changsha 410114, China.,Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Jianhui Wang
- Changsha University of Science & Technology, Changsha 410114, China
| | - Hongliang Zeng
- Research Institute of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha, Hunan 410013, China
| | - Fang Zhou
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Beibei Wen
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Xiangna Zhang
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Yong Luo
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Wenliang Wu
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Jianan Huang
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| | - Zhonghua Liu
- Key Laboratory of Ministry of Education for Tea Science, Hunan Agricultural University, Changsha 410128, China. .,National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients, Changsha 410128, China.
| |
Collapse
|
21
|
Zhang S, Dang Y. Roles of gut microbiota and metabolites in overweight and obesity of children. Front Endocrinol (Lausanne) 2022; 13:994930. [PMID: 36157438 PMCID: PMC9492854 DOI: 10.3389/fendo.2022.994930] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
The prevalence of overweight and obesity in children and adolescents is an increasing public health problem. Pediatric overweight and obesity result from multiple factors, including genetic background, diet, and lifestyle. In addition, the gut microbiota and their metabolites play crucial roles in the progression of overweight and obesity of children. Therefore, we reviewed the roles of gut microbiota in overweight/obese children. The relationship between pediatric overweight/obesity and gut metabolites, such as short-chain fatty acids, medium-chain fatty acids, amino acids, amines, and bile acids, are also summarized. Targeting gut microbiota and metabolites might be a promising strategy for interventions aimed at reducing pediatric overweight/obesity.
Collapse
Affiliation(s)
- Shengan Zhang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanqi Dang
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Yanqi Dang, ,
| |
Collapse
|
22
|
Dong TS, Guan M, Mayer EA, Stains J, Liu C, Vora P, Jacobs JP, Lagishetty V, Chang L, Barry RL, Gupta A. Obesity is associated with a distinct brain-gut microbiome signature that connects Prevotella and Bacteroides to the brain's reward center. Gut Microbes 2022; 14:2051999. [PMID: 35311453 PMCID: PMC8942409 DOI: 10.1080/19490976.2022.2051999] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 03/01/2022] [Indexed: 02/07/2023] Open
Abstract
The prevalence of obesity has risen to its highest values over the last two decades. While many studies have either shown brain or microbiome connections to obesity, few have attempted to analyze the brain-gut-microbiome relationship in a large cohort adjusting for cofounders. Therefore, we aim to explore the connection of the brain-gut-microbiome axis to obesity controlling for such cofounders as sex, race, and diet. Whole brain resting state functional MRI was acquired, and connectivity and brain network properties were calculated. Fecal samples were obtained from 287 obese and non-obese participants (males n = 99, females n = 198) for 16s rRNA profiling and fecal metabolites, along with a validated dietary questionnaire. Obesity was associated with alterations in the brain's reward network (nucleus accumbens, brainstem). Microbial diversity (p = .03) and composition (p = .03) differed by obesity independent of sex, race, or diet. Obesity was associated with an increase in Prevotella/Bacteroides (P/B) ratio and a decrease in fecal tryptophan (p = .02). P/B ratio was positively correlated to nucleus accumbens centrality (p = .03) and negatively correlated to fecal tryptophan (p = .004). Being Hispanic, eating a standard American diet, having a high Prevotella/Bacteroides ratio, and a high nucleus accumbens centrality were all independent risk factors for obesity. There are obesity-related signatures in the BGM-axis independent of sex, race, and diet. Race, diet, P/B ratio and increased nucleus accumbens centrality were independent risk factors for obesity. P/B ratio was inversely related to fecal tryptophan, a metabolite related to serotonin biosynthesis, and positively related to nucleus accumbens centrality, a region central to the brain's reward center. These findings may expand the field of therapies for obesity through novel pathways directed at the BGM axis.
Collapse
Affiliation(s)
- Tien S. Dong
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive DiseasesLos Angeles, USA
- Department of Medicine, David Geffen School of MedicineLos Angeles, USA
- Department of Medicine, UCLA Microbiome Center, David Geffen School of Medicine at UCLALos Angeles, USA
- Department of Medicine, G. Oppenheimer Center for Neurobiology of Stress and ResilienceLos Angeles, USA
- Department of Medicine, University of California, Los Angeles, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Michelle Guan
- Department of Medicine, David Geffen School of MedicineLos Angeles, USA
| | - Emeran A. Mayer
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive DiseasesLos Angeles, USA
- Department of Medicine, David Geffen School of MedicineLos Angeles, USA
- Department of Medicine, UCLA Microbiome Center, David Geffen School of Medicine at UCLALos Angeles, USA
- Department of Medicine, G. Oppenheimer Center for Neurobiology of Stress and ResilienceLos Angeles, USA
- Department of Medicine, University of California, Los Angeles, USA
| | - Jean Stains
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive DiseasesLos Angeles, USA
- Department of Medicine, David Geffen School of MedicineLos Angeles, USA
- Department of Medicine, G. Oppenheimer Center for Neurobiology of Stress and ResilienceLos Angeles, USA
- Department of Medicine, University of California, Los Angeles, USA
| | - Cathy Liu
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive DiseasesLos Angeles, USA
- Department of Medicine, David Geffen School of MedicineLos Angeles, USA
- Department of Medicine, G. Oppenheimer Center for Neurobiology of Stress and ResilienceLos Angeles, USA
- Department of Medicine, University of California, Los Angeles, USA
| | - Priten Vora
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive DiseasesLos Angeles, USA
- Department of Medicine, David Geffen School of MedicineLos Angeles, USA
- Department of Medicine, G. Oppenheimer Center for Neurobiology of Stress and ResilienceLos Angeles, USA
- Department of Medicine, University of California, Los Angeles, USA
| | - Jonathan P. Jacobs
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive DiseasesLos Angeles, USA
- Department of Medicine, UCLA Microbiome Center, David Geffen School of Medicine at UCLALos Angeles, USA
- Department of Medicine, University of California, Los Angeles, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Venu Lagishetty
- Department of Medicine, UCLA Microbiome Center, David Geffen School of Medicine at UCLALos Angeles, USA
- Department of Medicine, University of California, Los Angeles, USA
| | - Lin Chang
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive DiseasesLos Angeles, USA
- Department of Medicine, David Geffen School of MedicineLos Angeles, USA
- Department of Medicine, G. Oppenheimer Center for Neurobiology of Stress and ResilienceLos Angeles, USA
- Department of Medicine, University of California, Los Angeles, USA
| | - Robert L. Barry
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Harvard-Massachusetts Institute of Technology Health Sciences & Technology, Cambridge, MA, USA
| | - Arpana Gupta
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive DiseasesLos Angeles, USA
- Department of Medicine, David Geffen School of MedicineLos Angeles, USA
- Department of Medicine, UCLA Microbiome Center, David Geffen School of Medicine at UCLALos Angeles, USA
- Department of Medicine, G. Oppenheimer Center for Neurobiology of Stress and ResilienceLos Angeles, USA
- Department of Medicine, University of California, Los Angeles, USA
| |
Collapse
|
23
|
Plassmann H, Schelski DS, Simon M, Koban L. How we decide what to eat: Toward an interdisciplinary model of gut-brain interactions. WILEY INTERDISCIPLINARY REVIEWS. COGNITIVE SCIENCE 2022; 13:e1562. [PMID: 33977675 PMCID: PMC9286667 DOI: 10.1002/wcs.1562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 11/12/2022]
Abstract
Everyday dietary decisions have important short-term and long-term consequences for health and well-being. How do we decide what to eat, and what physiological and neurobiological systems are involved in those decisions? Here, we integrate findings from thus-far separate literatures: (a) the cognitive neuroscience of dietary decision-making, and (b) growing evidence of gut-brain interactions and especially influences of the gut microbiome on diet and health outcomes. We review findings that suggest that dietary decisions and food consumption influence nutrient sensing, homeostatic signaling in the gut, and the composition of the gut microbiome. In turn, the microbiome can influence host health and behavior. Through reward signaling pathways, the microbiome could potentially affect food and drink decisions. Such bidirectional links between gut microbiome and the brain systems underlying dietary decision-making may lead to self-reinforcing feedback loops that determine long-term dietary patterns, body mass, and health outcomes. This article is categorized under: Economics > Individual Decision-Making Psychology > Brain Function and Dysfunction Psychology > Reasoning and Decision Making.
Collapse
Affiliation(s)
- Hilke Plassmann
- Marketing AreaINSEADFontainebleauFrance
- Paris Brain Institute (ICM)INSERM U 1127, CNRS UMR 7225, Sorbonne UniversitéParisFrance
| | - Daniela Stephanie Schelski
- Center for Economics and NeuroscienceUniversity of BonnBonnGermany
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical CenterBonnGermany
| | - Marie‐Christine Simon
- Institute of Nutrition and Food Science (IEL), Nutrition and Microbiota, University of BonnBonnGermany
| | - Leonie Koban
- Marketing AreaINSEADFontainebleauFrance
- Paris Brain Institute (ICM)INSERM U 1127, CNRS UMR 7225, Sorbonne UniversitéParisFrance
| |
Collapse
|
24
|
Chernikova MA, Flores GD, Kilroy E, Labus JS, Mayer EA, Aziz-Zadeh L. The Brain-Gut-Microbiome System: Pathways and Implications for Autism Spectrum Disorder. Nutrients 2021; 13:nu13124497. [PMID: 34960049 PMCID: PMC8704412 DOI: 10.3390/nu13124497] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022] Open
Abstract
Gastrointestinal dysfunction is one of the most prevalent physiological symptoms of autism spectrum disorder (ASD). A growing body of largely preclinical research suggests that dysbiotic gut microbiota may modulate brain function and social behavior, yet little is known about the mechanisms that underlie these relationships and how they may influence the pathogenesis or severity of ASD. While various genetic and environmental risk factors have been implicated in ASD, this review aims to provide an overview of studies elucidating the mechanisms by which gut microbiota, associated metabolites, and the brain interact to influence behavior and ASD development, in at least a subgroup of individuals with gastrointestinal problems. Specifically, we review the brain-gut-microbiome system and discuss findings from current animal and human studies as they relate to social-behavioral and neurological impairments in ASD, microbiota-targeted therapies (i.e., probiotics, fecal microbiota transplantation) in ASD, and how microbiota may influence the brain at molecular, structural, and functional levels, with a particular interest in social and emotion-related brain networks. A deeper understanding of microbiome-brain-behavior interactions has the potential to inform new therapies aimed at modulating this system and alleviating both behavioral and physiological symptomatology in individuals with ASD.
Collapse
Affiliation(s)
- Michelle A. Chernikova
- USC Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA 90033, USA; (M.A.C.); (G.D.F.); (E.K.)
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA 90089, USA
- Psychology Department, Loyola Marymount University, Los Angeles, CA 90045, USA
| | - Genesis D. Flores
- USC Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA 90033, USA; (M.A.C.); (G.D.F.); (E.K.)
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA 90089, USA
- Psychology Department, California State Polytechnic University, Pomona, CA 91768, USA
| | - Emily Kilroy
- USC Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA 90033, USA; (M.A.C.); (G.D.F.); (E.K.)
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA 90089, USA
| | - Jennifer S. Labus
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California Los Angeles, Los Angeles, CA 90095, USA;
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA 90095, USA
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Gonda (Goldschmied) Neuroscience and Genetics Research Center, Brain Research Institute UCLA, Los Angeles, CA 90095, USA
| | - Emeran A. Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California Los Angeles, Los Angeles, CA 90095, USA;
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA 90095, USA
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Correspondence: (E.A.M.); (L.A.-Z.)
| | - Lisa Aziz-Zadeh
- USC Chan Division of Occupational Science and Occupational Therapy, University of Southern California, Los Angeles, CA 90033, USA; (M.A.C.); (G.D.F.); (E.K.)
- Brain and Creativity Institute, University of Southern California, Los Angeles, CA 90089, USA
- Correspondence: (E.A.M.); (L.A.-Z.)
| |
Collapse
|
25
|
Labus JS, Mayer EA, Tillisch K, Aagaard KM, Stains J, Broniowska K, Van Remortel C, Tun G, Rapkin A. Dysregulation in Sphingolipid Signaling Pathways is Associated With Symptoms and Functional Connectivity of Pain Processing Brain Regions in Provoked Vestibulodynia. THE JOURNAL OF PAIN 2021; 22:1586-1605. [PMID: 34029688 PMCID: PMC10460622 DOI: 10.1016/j.jpain.2021.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/27/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
Provoked vestibulodynia (PVD) is a chronic pain disorder characterized by local hypersensitivity and severe pain with pressure localized to the vulvar vestibule. Despite decades of study, the lack of identified biomarkers has slowed the development of effective therapies. The primary aim of this study was to use metabolomics to identify novel biochemical mechanisms in vagina and blood underlying brain biomarkers and symptoms in PVD, thereby closing this knowledge gap. Using a cross-sectional case-control observational study design, untargeted and unbiased metabolomic profiling of vaginal fluid and plasma was performed in women with PVD compared to healthy controls. In women with PVD, we also obtained assessments of vulvar pain, vestibular and vaginal muscle tenderness, and 24-hour symptom intensity alongside resting-state brain functional connectivity of brain regions involved in pain processing and modulation. Compared to healthy controls, women with PVD demonstrated differences primarily in vaginal (but not plasma) concentrations of metabolites of the sphingolipid signaling pathways, suggesting localized effects in vagina and vulvar vestibule rather than systemic effects. Our findings reveal that dysregulation of sphingolipid metabolism in PVD is associated with increased vulvar pain and muscle tenderness, sexual dysfunction, and decreased functional connectivity strength in pain processing/modulatory brain regions. This data collectively suggests that alterations in sphingolipid signaling pathways are likely an important molecular biomarker in PVD that could lead to new targets for therapeutic intervention. PERSPECTIVE: This manuscript presents the results of a robust, unbiased molecular assessment of plasma and vaginal fluid samples in women with provoked vestibulodynia compared to healthy controls. The findings suggest that alterations in sphingolipid signaling pathways are associated with symptoms and brain biomarkers and may be an important molecular marker that could provide new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jennifer S Labus
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California; Brain Research Institute UCLA, Gonda (Goldschmied) Neuroscience and Genetics Research Center, Los Angeles, California.
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Kirsten Tillisch
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Kjersti M Aagaard
- Division of Maternal-Fetal Medicine, Departments of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas; Department of Molecular and Human Genetics, Bioinformatics Research Laboratory, Baylor College of Medicine, Houston, Texas; Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, Texas
| | - Jean Stains
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | | | - Charlotte Van Remortel
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Guistinna Tun
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California
| | - Andrea Rapkin
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, California; Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California, Los Angeles, California
| |
Collapse
|
26
|
Barone M, D'Amico F, Fabbrini M, Rampelli S, Brigidi P, Turroni S. Over-feeding the gut microbiome: A scoping review on health implications and therapeutic perspectives. World J Gastroenterol 2021; 27:7041-7064. [PMID: 34887627 PMCID: PMC8613651 DOI: 10.3748/wjg.v27.i41.7041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/02/2021] [Accepted: 10/14/2021] [Indexed: 02/06/2023] Open
Abstract
The human gut microbiome has gained increasing attention over the past two decades. Several findings have shown that this complex and dynamic microbial ecosystem can contribute to the maintenance of host health or, when subject to imbalances, to the pathogenesis of various enteric and non-enteric diseases. This scoping review summarizes the current knowledge on how the gut microbiota and microbially-derived compounds affect host metabolism, especially in the context of obesity and related disorders. Examples of microbiome-based targeted intervention strategies that aim to restore and maintain an eubiotic layout are then discussed. Adjuvant therapeutic interventions to alleviate obesity and associated comorbidities are traditionally based on diet modulation and the supplementation of prebiotics, probiotics and synbiotics. However, these approaches have shown only moderate ability to induce sustained changes in the gut microbial ecosystem, making the development of innovative and tailored microbiome-based intervention strategies of utmost importance in clinical practice. In this regard, the administration of next-generation probiotics and engineered microbiomes has shown promising results, together with more radical intervention strategies based on the replacement of the dysbiotic ecosystem by means of fecal microbiota transplantation from healthy donors or with the introduction of synthetic communities specifically designed to achieve the desired therapeutic outcome. Finally, we provide a perspective for future translational investigations through the implementation of bioinformatics approaches, including machine and deep learning, to predict health risks and therapeutic outcomes.
Collapse
Affiliation(s)
- Monica Barone
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Federica D'Amico
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Marco Fabbrini
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Simone Rampelli
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| | - Patrizia Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna 40138, Italy
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40126, Italy
| |
Collapse
|
27
|
Coley EJ, Mayer EA, Osadchiy V, Chen Z, Subramanyam V, Zhang Y, Hsiao EY, Gao K, Bhatt R, Dong T, Vora P, Naliboff B, Jacobs JP, Gupta A. Early life adversity predicts brain-gut alterations associated with increased stress and mood. Neurobiol Stress 2021; 15:100348. [PMID: 34113697 PMCID: PMC8170500 DOI: 10.1016/j.ynstr.2021.100348] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 01/16/2023] Open
Abstract
Alterations in the brain-gut system have been implicated in various disease states, but little is known about how early-life adversity (ELA) impacts development and adult health as mediated by brain-gut interactions. We hypothesize that ELA disrupts components of the brain-gut system, thereby increasing susceptibility to disordered mood. In a sample of 128 healthy adult participants, a history of ELA and current stress, depression, and anxiety were assessed using validated questionnaires. Fecal metabolites were measured using liquid chromatography tandem mass spectrometry-based untargeted metabolomic profiling. Functional brain connectivity was evaluated by magnetic resonance imaging. Sparse partial least squares-discriminant analysis, controlling for sex, body mass index, age, and diet was used to predict brain-gut alterations as a function of ELA. ELA was correlated with four gut-regulated metabolites within the glutamate pathway (5-oxoproline, malate, urate, and glutamate gamma methyl ester) and alterations in functional brain connectivity within primarily sensorimotor, salience, and central executive networks. Integrated analyses revealed significant associations between these metabolites, functional brain connectivity, and scores for perceived stress, anxiety, and depression. This study reveals a novel association between a history of ELA, alterations in the brain-gut axis, and increased vulnerability to negative mood and stress. Results from the study raise the hypothesis that select gut-regulated metabolites may contribute to the adverse effects of critical period stress on neural development via pathways related to glutamatergic excitotoxicity and oxidative stress.
Collapse
Affiliation(s)
- Elena J.L. Coley
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Emeran A. Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, USA,David Geffen School of Medicine, University of California, Los Angeles, USA,Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, CA, USA,UCLA Microbiome Center, Los Angeles, CA, USA
| | - Vadim Osadchiy
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, USA,David Geffen School of Medicine, University of California, Los Angeles, USA,Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zixi Chen
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, USA
| | - Vishvak Subramanyam
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, USA
| | - Yurui Zhang
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, USA
| | - Elaine Y. Hsiao
- David Geffen School of Medicine, University of California, Los Angeles, USA,UCLA Microbiome Center, Los Angeles, CA, USA,Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kan Gao
- Laboratory of Gastrointestinal Microbiology, Nanjing Agricultural University, PR China
| | - Ravi Bhatt
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, USA,Imaging Genetics Center, Institute for Neuroimaging and Informatics, University of Southern California, Los Angeles, CA, USA
| | - Tien Dong
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, USA,David Geffen School of Medicine, University of California, Los Angeles, USA,Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, CA, USA,UCLA Microbiome Center, Los Angeles, CA, USA,Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Priten Vora
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, USA
| | - Bruce Naliboff
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, USA,David Geffen School of Medicine, University of California, Los Angeles, USA,Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, CA, USA
| | - Jonathan P. Jacobs
- David Geffen School of Medicine, University of California, Los Angeles, USA,Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, CA, USA,UCLA Microbiome Center, Los Angeles, CA, USA,Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Arpana Gupta
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, USA,David Geffen School of Medicine, University of California, Los Angeles, USA,Vatche and Tamar Manoukian Division of Digestive Diseases, Los Angeles, CA, USA,UCLA Microbiome Center, Los Angeles, CA, USA,Corresponding author. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA CHS, 42-210 MC737818 10833 Le Conte Avenue, USA.
| |
Collapse
|
28
|
Abstract
The gut microbiota has the capacity to affect host appetite via intestinal satiety pathways, as well as complex feeding behaviors. In this Review, we highlight recent evidence that the gut microbiota can modulate food preference across model organisms. We discuss effects of the gut microbiota on the vagus nerve and brain regions including the hypothalamus, mesolimbic system, and prefrontal cortex, which play key roles in regulating feeding behavior. Crosstalk between commensal bacteria and the central and peripheral nervous systems is associated with alterations in signaling of neurotransmitters and neuropeptides such as dopamine, brain-derived neurotrophic factor (BDNF), and glucagon-like peptide-1 (GLP-1). We further consider areas for future research on mechanisms by which gut microbes may influence feeding behavior involving these neural pathways. Understanding roles for the gut microbiota in feeding regulation will be important for informing therapeutic strategies to treat metabolic and eating disorders.
Collapse
|
29
|
Food addiction and psychiatric comorbidities: a review of current evidence. Eat Weight Disord 2021; 26:1049-1056. [PMID: 32968944 DOI: 10.1007/s40519-020-01021-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/12/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Food addiction (FA) is characterised by the consumption of appetible foods and by addictive psychological and behavioural symptoms such as cravings, tolerance, limited control of substance intake and withdrawal symptoms. Despite previous research on FA has been hindered by the lack of a formal definition for this condition, recent global trends have stirred the interest of the scientific community towards a proper classification and construct of FA. More specifically, recent studies have pointed towards shared defective neurobiological mechanisms as well as frequent comorbidities between FA, eating disorders, mood disorders, anxiety disorders and substance-related and addictive disorders. OBJECTIVE In this review, we will provide an overview of the complex symptomatology of food addiction evaluating its relationship with mood disorders, anxiety disorders, eating disorders and substance-related and addictive disorders. METHODS We wrote a systematic review and followed a PRISMA methods. RESULTS Patients with FA and substance use disorders show similar risk factors, neurobiological and hormonal correlates, personality traits and symptom profiles. The presence of FA appears to be directly proportional to the burden of symptoms of affective disorder. The comorbidity between FA and other eating disorders is associated with worse clinical conditions and symptoms. CONCLUSION FA should be considered a sort of transnosological construct existing in different psychopathological domains that have similarities with substance-related, affective, and eating disorders. Furthermore, FA seems to be likely an important factor related to several psychopathological dimensions, but further studies are needed to clarify this view. LEVEL OF EVIDENCE Level V, review article.
Collapse
|
30
|
Margolis KG, Cryan JF, Mayer EA. The Microbiota-Gut-Brain Axis: From Motility to Mood. Gastroenterology 2021; 160:1486-1501. [PMID: 33493503 PMCID: PMC8634751 DOI: 10.1053/j.gastro.2020.10.066] [Citation(s) in RCA: 385] [Impact Index Per Article: 128.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/07/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023]
Abstract
The gut-brain axis plays an important role in maintaining homeostasis. Many intrinsic and extrinsic factors influence signaling along this axis, modulating the function of both the enteric and central nervous systems. More recently the role of the microbiome as an important factor in modulating gut-brain signaling has emerged and the concept of a microbiota-gut-brain axis has been established. In this review, we highlight the role of this axis in modulating enteric and central nervous system function and how this may impact disorders such as irritable bowel syndrome and disorders of mood and affect. We examine the overlapping biological constructs that underpin these disorders with a special emphasis on the neurotransmitter serotonin, which plays a key role in both the gastrointestinal tract and in the brain. Overall, it is clear that although animal studies have shown much promise, more progress is necessary before these findings can be translated for diagnostic and therapeutic benefit in patient populations.
Collapse
Affiliation(s)
- Kara G. Margolis
- Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University Irving Medical Center, New York, NY,Corresponding author:
| | - John F. Cryan
- Department of Anatomy & Neuroscience, University College Cork, Ireland, APC Microbiome Ireland, University College Cork, Ireland
| | - Emeran A. Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vachte and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
31
|
Lai Y, Liu CW, Chi L, Ru H, Lu K. High-Resolution Metabolomics of 50 Neurotransmitters and Tryptophan Metabolites in Feces, Serum, and Brain Tissues Using UHPLC-ESI-Q Exactive Mass Spectrometry. ACS OMEGA 2021; 6:8094-8103. [PMID: 33817468 PMCID: PMC8014936 DOI: 10.1021/acsomega.0c05789] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/25/2021] [Indexed: 05/31/2023]
Abstract
Recent evidence indicates that tryptophan metabolites and neurotransmitters are potential mediators of the microbiome-gut-brain interaction. Here, a high-resolution ultra-high performance liquid chromatography-electrospray ionization tandem mass spectrometry (UHPLC-ESI-MS/MS) assay was developed and validated for quantifying 50 neurotransmitters, tryptophan metabolites, and bacterial indole derivatives in mouse serum, feces, and brain. The lower limit of quantitation for the 50 compounds ranged from 0.5 to 100 nmol/L, and sample preparation procedures were adapted for individual compounds to allow quantitation within linearity of the assay with a correlation coefficient >0.99. Reproducibility was tested by intra- and interday precision and accuracy of analysis: intra- and interday precision at the lower limit of quantitation was less than 20% for all compounds, with over two-thirds of the compounds achieving an interday precision below 10%, while the interday accuracy at the lower limit of quantitation ranged from 82.3 to 128.0% for all compounds. The analyte recovery was assessed based on sample-spiked stable-isotope-labeling standards, illustrating a need to consider matrix-specific recovery discrepancies when performing interorgan comparison. Carryover was evaluated by intermittent solvent blank injection. The assay was successfully applied to determining the concentration profiles of neurotransmitter and tryptophan metabolites in serum, feces, and brain of conventionally raised specific pathogen-free (SPF) C57BL/6 mice. Our method may serve as a useful analytical resource for investigating the roles of tryptophan metabolism and neurotransmitter signaling in host-microbiota interaction.
Collapse
|
32
|
Constante M, De Palma G, Lu J, Jury J, Rondeau L, Caminero A, Collins SM, Verdu EF, Bercik P. Saccharomyces boulardii CNCM I-745 modulates the microbiota-gut-brain axis in a humanized mouse model of Irritable Bowel Syndrome. Neurogastroenterol Motil 2021; 33:e13985. [PMID: 32955166 DOI: 10.1111/nmo.13985] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Gnotobiotic mice colonized with microbiota from patients with irritable bowel syndrome (IBS) and comorbid anxiety (IBS+A) display gut dysfunction and anxiety-like behavior compared to mice colonized with microbiota from healthy volunteers. Using this model, we tested the therapeutic potential of the probiotic yeast Saccharomyces boulardii strain CNCM I-745 (S. bou) and investigated underlying mechanisms. METHODS Germ-free Swiss Webster mice were colonized with fecal microbiota from an IBS+A patient or a healthy control (HC). Three weeks later, mice were gavaged daily with S. boulardii or placebo for two weeks. Anxiety-like behavior (light preference and step-down tests), gastrointestinal transit, and permeability were assessed. After sacrifice, samples were taken for gene expression by NanoString and qRT-PCR, microbiota 16S rRNA profiling, and indole quantification. KEY RESULTS Mice colonized with IBS+A microbiota developed faster gastrointestinal transit and anxiety-like behavior (longer step-down latency) compared to mice with HC microbiota. S. bou administration normalized gastrointestinal transit and anxiety-like behavior in mice with IBS+A microbiota. Step-down latency correlated with colonic Trpv1 expression and was associated with altered microbiota profile and increased Indole-3-acetic acid (IAA) levels. CONCLUSIONS & INFERENCES Treatment with S. bou improves gastrointestinal motility and anxiety-like behavior in mice with IBS+A microbiota. Putative mechanisms include effects on pain pathways, direct modulation of the microbiota, and indole production by commensal bacteria.
Collapse
Affiliation(s)
- Marco Constante
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Giada De Palma
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Jun Lu
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Jennifer Jury
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Liam Rondeau
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Alberto Caminero
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Stephen M Collins
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Premysl Bercik
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
33
|
Food Addiction and Tobacco Use Disorder: Common Liability and Shared Mechanisms. Nutrients 2020; 12:nu12123834. [PMID: 33334010 PMCID: PMC7765398 DOI: 10.3390/nu12123834] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/04/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
As food addiction is being more commonly recognized within the scientific community, parallels can be drawn between it and other addictive substance use disorders, including tobacco use disorder. Given that both unhealthy diets and smoking are leading risk factors for disability and death, a greater understanding of how food addiction and tobacco use disorder overlap with one another is necessary. This narrative review aimed to highlight literature that investigated prevalence, biology, psychology, and treatment options of food addiction and tobacco use disorder. Published studies up to August 2020 and written in English were included. Using a biopsychosocial lens, each disorder was assessed together and separately, as there is emerging evidence that the two disorders can develop concurrently or sequentially within individuals. Commonalities include but are not limited to the dopaminergic neurocircuitry, gut microbiota, childhood adversity, and attachment insecurity. In addition, the authors conducted a feasibility study with the purpose of examining the association between food addiction symptoms and tobacco use disorder among individuals seeking tobacco use disorder treatment. To inform future treatment approaches, more research is necessary to identify and understand the overlap between the two disorders.
Collapse
|
34
|
Longitudinal Changes in Food Addiction Symptoms and Body Weight among Adults in a Behavioral Weight-Loss Program. Nutrients 2020; 12:nu12123687. [PMID: 33260468 PMCID: PMC7760227 DOI: 10.3390/nu12123687] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Interest in food addiction (FA) has increased, but little is known about its clinical implications or potential treatments. Using secondary analyses from a randomized controlled trial, we evaluated the associations between changes in FA, body weight, and “problem food” consumption during a 22-month behavioral weight-loss program consisting of an initial four-month in-person intervention, 12-month extended-care, and six-month follow-up (n = 182). Food addiction was measured using the Yale Food Addiction Scale. “Problem foods” were identified from the literature and self-reporting. Multilevel modeling was used as the primary method of analysis. We hypothesized that reductions in problem food consumption during the initial treatment phase would be associated with long-term (22-month) FA reductions. As expected, we found that reductions in problem foods were associated with greater initial reductions in FA symptoms; however, they were also associated with a sharper rebound in symptoms over time (p = 0.016), resulting in no significant difference at Month 22 (p = 0.856). Next, we hypothesized that long-term changes in FA would be associated with long-term changes in body weight. Although both FA and weight decreased over time (ps < 0.05), month-to-month changes in FA were not associated with month-to-month changes in weight (p = 0.706). Instead, higher overall FA (i.e., mean scores over the course of the study) were associated with less weight loss (p = 0.008) over time. Finally, we hypothesized that initial reductions in problem food consumption would be associated with long-term reductions in weight, but this relationship was not significant (ps > 0.05). Given the complexity of the findings, more research is needed to identify interventions for long-term changes in FA and to elucidate the associations between problem foods, FA, and weight.
Collapse
|
35
|
Constant A, Moirand R, Thibault R, Val-Laillet D. Meeting of Minds around Food Addiction: Insights from Addiction Medicine, Nutrition, Psychology, and Neurosciences. Nutrients 2020; 12:nu12113564. [PMID: 33233694 PMCID: PMC7699750 DOI: 10.3390/nu12113564] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/12/2020] [Accepted: 11/18/2020] [Indexed: 12/21/2022] Open
Abstract
This review, focused on food addiction (FA), considers opinions from specialists with different expertise in addiction medicine, nutrition, health psychology, and behavioral neurosciences. The concept of FA is a recurring issue in the clinical description of abnormal eating. Even though some tools have been developed to diagnose FA, such as the Yale Food Addiction Scale (YFAS) questionnaire, the FA concept is not recognized as an eating disorder (ED) so far and is even not mentioned in the Diagnostic and Statistical Manuel of Mental Disorders version 5 (DSM-5) or the International Classification of Disease (ICD-11). Its triggering mechanisms and relationships with other substance use disorders (SUD) need to be further explored. Food addiction (FA) is frequent in the overweight or obese population, but it remains unclear whether it could articulate with obesity-related comorbidities. As there is currently no validated therapy against FA in obese patients, FA is often underdiagnosed and untreated, so that FA may partly explain failure of obesity treatment, addiction transfer, and weight regain after obesity surgery. Future studies should assess whether a dedicated management of FA is associated with better outcomes, especially after obesity surgery. For prevention and treatment purposes, it is necessary to promote a comprehensive psychological approach to FA. Understanding the developmental process of FA and identifying precociously some high-risk profiles can be achieved via the exploration of the environmental, emotional, and cognitive components of eating, as well as their relationships with emotion management, some personality traits, and internalized weight stigma. Under the light of behavioral neurosciences and neuroimaging, FA reveals a specific brain phenotype that is characterized by anomalies in the reward and inhibitory control processes. These anomalies are likely to disrupt the emotional, cognitive, and attentional spheres, but further research is needed to disentangle their complex relationship and overlap with obesity and other forms of SUD. Prevention, diagnosis, and treatment must rely on a multidisciplinary coherence to adapt existing strategies to FA management and to provide social and emotional support to these patients suffering from highly stigmatized medical conditions, namely overweight and addiction. Multi-level interventions could combine motivational interviews, cognitive behavioral therapies, and self-help groups, while benefiting from modern exploratory and interventional tools to target specific neurocognitive processes.
Collapse
Affiliation(s)
- Aymery Constant
- INRAE, INSERM, University Rennes, NuMeCan, Nutrition Metabolisms Cancer, 35590 St Gilles, 35000 Rennes, France; (A.C.); (R.M.); (R.T.)
- EHESP, School of Public Health, 35043 Rennes, France
| | - Romain Moirand
- INRAE, INSERM, University Rennes, NuMeCan, Nutrition Metabolisms Cancer, 35590 St Gilles, 35000 Rennes, France; (A.C.); (R.M.); (R.T.)
- Unité d’Addictologie, CHU Rennes, 35000 Rennes, France
| | - Ronan Thibault
- INRAE, INSERM, University Rennes, NuMeCan, Nutrition Metabolisms Cancer, 35590 St Gilles, 35000 Rennes, France; (A.C.); (R.M.); (R.T.)
- Unité de Nutrition, CHU Rennes, 35000 Rennes, France
| | - David Val-Laillet
- INRAE, INSERM, University Rennes, NuMeCan, Nutrition Metabolisms Cancer, 35590 St Gilles, 35000 Rennes, France; (A.C.); (R.M.); (R.T.)
- Correspondence:
| |
Collapse
|
36
|
Osadchiy V, Mayer EA, Gao K, Labus JS, Naliboff B, Tillisch K, Chang L, Jacobs JP, Hsiao EY, Gupta A. Analysis of brain networks and fecal metabolites reveals brain-gut alterations in premenopausal females with irritable bowel syndrome. Transl Psychiatry 2020; 10:367. [PMID: 33139708 PMCID: PMC7608552 DOI: 10.1038/s41398-020-01071-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/11/2020] [Accepted: 10/05/2020] [Indexed: 01/16/2023] Open
Abstract
Alterations in brain-gut-microbiome (BGM) interactions have been implicated in the pathogenesis of irritable bowel syndrome (IBS). Here, we apply a systems biology approach, leveraging neuroimaging and fecal metabolite data, to characterize BGM interactions that are driving IBS pathophysiology. Fecal samples and resting state fMRI images were obtained from 138 female subjects (99 IBS, 39 healthy controls (HCs)). Partial least-squares discriminant analysis (PLS-DA) was conducted to explore group differences, and partial correlation analysis explored significantly changed metabolites and neuroimaging data. All correlational tests were performed controlling for age, body mass index, and diet; results are reported after FDR correction, with q < 0.05 as significant. Compared to HCs, IBS showed increased connectivity of the putamen with regions of the default mode and somatosensory networks. Metabolite pathways involved in nucleic acid and amino acid metabolism differentiated the two groups. Only a subset of metabolites, primarily amino acids, were associated with IBS-specific brain changes, including tryptophan, glutamate, and histidine. Histidine was the only metabolite positively associated with both IBS-specific alterations in brain connectivity. Our findings suggest a role for several amino acid metabolites in modulating brain function in IBS. These metabolites may alter brain connectivity directly, by crossing the blood-brain-barrier, or indirectly through peripheral mechanisms. This is the first study to integrate both neuroimaging and fecal metabolite data supporting the BGM model of IBS, building the foundation for future mechanistic studies on the influence of gut microbial metabolites on brain function in IBS.
Collapse
Affiliation(s)
- Vadim Osadchiy
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- UCLA Microbiome Center, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Kan Gao
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jennifer S Labus
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Bruce Naliboff
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Kirsten Tillisch
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- UCLA Microbiome Center, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Lin Chang
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Jonathan P Jacobs
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- UCLA Microbiome Center, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Elaine Y Hsiao
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Arpana Gupta
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California, Los Angeles, Los Angeles, CA, USA.
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Vatche and Tamar Manoukian Division of Digestive Diseases, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA.
| |
Collapse
|
37
|
Gupta A, Osadchiy V, Mayer EA. Brain-gut-microbiome interactions in obesity and food addiction. Nat Rev Gastroenterol Hepatol 2020; 17:655-672. [PMID: 32855515 PMCID: PMC7841622 DOI: 10.1038/s41575-020-0341-5] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/24/2020] [Indexed: 12/13/2022]
Abstract
Normal eating behaviour is coordinated by the tightly regulated balance between intestinal and extra-intestinal homeostatic and hedonic mechanisms. By contrast, food addiction is a complex, maladaptive eating behaviour that reflects alterations in brain-gut-microbiome (BGM) interactions and a shift of this balance towards hedonic mechanisms. Each component of the BGM axis has been implicated in the development of food addiction, with both brain to gut and gut to brain signalling playing a role. Early-life influences can prime the infant gut microbiome and brain for food addiction, which might be further reinforced by increased antibiotic usage and dietary patterns throughout adulthood. The ubiquitous availability and marketing of inexpensive, highly palatable and calorie-dense food can further shift this balance towards hedonic eating through both central (disruptions in dopaminergic signalling) and intestinal (vagal afferent function, metabolic endotoxaemia, systemic immune activation, changes to gut microbiome and metabolome) mechanisms. In this Review, we propose a systems biology model of BGM interactions, which incorporates published reports on food addiction, and provides novel insights into treatment targets aimed at each level of the BGM axis.
Collapse
Affiliation(s)
- Arpana Gupta
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity Program, University of California Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA, USA
| | - Vadim Osadchiy
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity Program, University of California Los Angeles, Los Angeles, CA, USA
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Emeran A Mayer
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity Program, University of California Los Angeles, Los Angeles, CA, USA.
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA, USA.
- Ahmanson-Lovelace Brain Mapping Center at University of California Los Angeles, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
38
|
Sarkar A, Harty S, Johnson KVA, Moeller AH, Carmody RN, Lehto SM, Erdman SE, Dunbar RIM, Burnet PWJ. The role of the microbiome in the neurobiology of social behaviour. Biol Rev Camb Philos Soc 2020; 95:1131-1166. [PMID: 32383208 PMCID: PMC10040264 DOI: 10.1111/brv.12603] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022]
Abstract
Microbes colonise all multicellular life, and the gut microbiome has been shown to influence a range of host physiological and behavioural phenotypes. One of the most intriguing and least understood of these influences lies in the domain of the microbiome's interactions with host social behaviour, with new evidence revealing that the gut microbiome makes important contributions to animal sociality. However, little is known about the biological processes through which the microbiome might influence host social behaviour. Here, we synthesise evidence of the gut microbiome's interactions with various aspects of host sociality, including sociability, social cognition, social stress, and autism. We discuss evidence of microbial associations with the most likely physiological mediators of animal social interaction. These include the structure and function of regions of the 'social' brain (the amygdala, the prefrontal cortex, and the hippocampus) and the regulation of 'social' signalling molecules (glucocorticoids including corticosterone and cortisol, sex hormones including testosterone, oestrogens, and progestogens, neuropeptide hormones such as oxytocin and arginine vasopressin, and monoamine neurotransmitters such as serotonin and dopamine). We also discuss microbiome-associated host genetic and epigenetic processes relevant to social behaviour. We then review research on microbial interactions with olfaction in insects and mammals, which contribute to social signalling and communication. Following these discussions, we examine evidence of microbial associations with emotion and social behaviour in humans, focussing on psychobiotic studies, microbe-depression correlations, early human development, autism, and issues of statistical power, replication, and causality. We analyse how the putative physiological mediators of the microbiome-sociality connection may be investigated, and discuss issues relating to the interpretation of results. We also suggest that other candidate molecules should be studied, insofar as they exert effects on social behaviour and are known to interact with the microbiome. Finally, we consider different models of the sequence of microbial effects on host physiological development, and how these may contribute to host social behaviour.
Collapse
Affiliation(s)
- Amar Sarkar
- Trinity College, Trinity Street, University of Cambridge, Cambridge, CB2 1TQ, U.K.,Leverhulme Centre for Human Evolutionary Studies, Department of Archaeology, Fitzwilliam Street, University of Cambridge, Cambridge, CB2 1QH, U.K
| | - Siobhán Harty
- Institute of Neuroscience, Trinity College Dublin, Dublin 2, Dublin, Ireland.,School of Psychology, Trinity College Dublin, Dublin 2, Dublin, Ireland
| | - Katerina V-A Johnson
- Department of Experimental Psychology, Radcliffe Observatory Quarter, University of Oxford, Oxford, OX2 6GG, U.K.,Pembroke College, University of Oxford, Oxford, OX1 1DW, U.K.,Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX, U.K
| | - Andrew H Moeller
- Department of Ecology and Evolutionary Biology, Corson Hall, Tower Road, Cornell University, Ithaca, NY, 14853, U.S.A
| | - Rachel N Carmody
- Department of Human Evolutionary Biology, Harvard University, Peabody Museum, 11 Divinity Avenue, Cambridge, Massachusetts, 02138, USA
| | - Soili M Lehto
- Psychiatry, University of Helsinki and Helsinki University Hospital, PL 590, FI-00029, Helsinki, Finland.,Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, P.O. Box 6, FI-00014, Helsinki, Finland.,Institute of Clinical Medicine/Psychiatry, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Susan E Erdman
- Division of Comparative Medicine, Massachusetts Institute of Technology, Building 16-825, 77 Massachusetts Avenue, Cambridge, MA, 02139, U.S.A
| | - Robin I M Dunbar
- Department of Experimental Psychology, Radcliffe Observatory Quarter, University of Oxford, Oxford, OX2 6GG, U.K
| | - Philip W J Burnet
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX, U.K
| |
Collapse
|
39
|
Effects of the Human Gut Microbiota on Cognitive Performance, Brain Structure and Function: A Narrative Review. Nutrients 2020; 12:nu12103009. [PMID: 33007941 PMCID: PMC7601389 DOI: 10.3390/nu12103009] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
Enhancing or preserving cognitive performance of personnel working in stressful, demanding and/or high tempo environments is vital for optimal performance. Emerging research suggests that the human gut microbiota may provide a potential avenue to enhance cognition. This review examines the relationship between the human gut microbiota, including modulators of the microbiota on cognition and/or brain function. For this narrative review, a total of n = 17 relevant human research items of a possible 1765 published between January 2010 and November 2018 were identified. Two overarching design methods for synthesis were observed: correlational or pre/post intervention. Limited correlational design studies linking microbiota to cognitive/brain structure endpoints existed (n = 5); however, correlations between microbiota diversity and enhanced cognitive flexibility and executive function were observed. Gut microbiota intervention studies to improve cognition or brain function (n = 12) generally resulted in improved cognition (11/12), in which improvements were observed in visuospatial memory, verbal learning and memory, and aspects of attentional vigilance. Limited studies were available to draw a detailed conclusion; however, available evidence suggests that gut microbiota is linked to cognitive performance and that manipulation of gut microbiota could be a promising avenue for enhancing cognition which warrants further research.
Collapse
|
40
|
Ma N, He T, Johnston LJ, Ma X. Host-microbiome interactions: the aryl hydrocarbon receptor as a critical node in tryptophan metabolites to brain signaling. Gut Microbes 2020; 11:1203-1219. [PMID: 32401136 PMCID: PMC7524279 DOI: 10.1080/19490976.2020.1758008] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tryptophan (Trp) is not only a nutrient enhancer but also has systemic effects. Trp metabolites signaling through the well-known aryl hydrocarbon receptor (AhR) constitute the interface of microbiome-gut-brain axis. However, the pathway through which Trp metabolites affect central nervous system (CNS) function have not been fully elucidated. AhR participates in a broad variety of physiological and pathological processes that also highly relevant to intestinal homeostasis and CNS diseases. Via the AhR-dependent mechanism, Trp metabolites connect bidirectional signaling between the gut microbiome and the brain, mediated via immune, metabolic, and neural (vagal) signaling mechanisms, with downstream effects on behavior and CNS function. These findings shed light on the complex Trp regulation of microbiome-gut-brain axis and add another facet to our understanding that dietary Trp is expected to be a promising noninvasive approach for alleviating systemic diseases.
Collapse
Affiliation(s)
- Ning Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ting He
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lee J. Johnston
- West Central Research & Outreach Center, University of Minnesota, Morris, MN, USA
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China,CONTACT Xi Ma State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2, Yuanmingyuan West Road, Haidian District, Beijing100193, China
| |
Collapse
|
41
|
Dong TS, Mayer EA, Osadchiy V, Chang C, Katzka W, Lagishetty V, Gonzalez K, Kalani A, Stains J, Jacobs JP, Longo VD, Gupta A. A Distinct Brain-Gut-Microbiome Profile Exists for Females with Obesity and Food Addiction. Obesity (Silver Spring) 2020; 28:1477-1486. [PMID: 32935533 PMCID: PMC7494955 DOI: 10.1002/oby.22870] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/07/2020] [Accepted: 04/21/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Alterations in brain-gut-microbiome interactions have been implicated as an important factor in obesity. This study aimed to explore the relationship between food addiction (FA) and the brain-gut-microbiome axis, using a multi-omics approach involving microbiome data, metabolomics, and brain imaging. METHODS Brain magnetic resonance imaging was obtained in 105 females. FA was defined by using the Yale Food Addiction Scale. Fecal samples were collected for sequencing and metabolomics. Statistical analysis was done by using multivariate analyses and machine learning algorithms. RESULTS Of the females with obesity, 33.3% exhibited FA as compared with 5.3% and 0.0% of females with overweight and normal BMI, respectively (P = 0.0001). Based on a multilevel sparse partial least square discriminant analysis, there was a difference in the gut microbiome of females with FA versus those without. Differential abundance testing showed Bacteroides, Megamonas, Eubacterium, and Akkermansia were statistically associated with FA (q < 0.05). Metabolomics showed that indolepropionic acid was inversely correlated with FA. FA was also correlated with increased connectivity within the brain's reward network, specifically between the intraparietal sulcus, brain stem, and putamen. CONCLUSIONS This is the first study to examine FA along the brain-gut-microbiome axis and it supports the idea of targeting the brain-gut-microbiome axis for the treatment of FA and obesity.
Collapse
Affiliation(s)
- Tien S. Dong
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- David Geffen School of Medicine, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Emeran A. Mayer
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- David Geffen School of Medicine, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Vadim Osadchiy
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Candace Chang
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - William Katzka
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Venu Lagishetty
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Kimberly Gonzalez
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Amir Kalani
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Jean Stains
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Jonathan P. Jacobs
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- David Geffen School of Medicine, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| | - Valter D. Longo
- USC Longevity Institute, University of Southern California, Los Angeles
| | - Arpana Gupta
- Vatche and Tamar Manoukian Division of Digestive Diseases, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- David Geffen School of Medicine, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- UCLA Microbiome Center, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
- University of California, Los Angeles, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA
| |
Collapse
|
42
|
Tabibnia G. An affective neuroscience model of boosting resilience in adults. Neurosci Biobehav Rev 2020; 115:321-350. [DOI: 10.1016/j.neubiorev.2020.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 05/09/2020] [Accepted: 05/10/2020] [Indexed: 12/11/2022]
|
43
|
Potential therapeutic applications of the gut microbiome in obesity: from brain function to body detoxification. Int J Obes (Lond) 2020; 44:1818-1831. [PMID: 32523034 DOI: 10.1038/s41366-020-0618-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/04/2020] [Accepted: 05/22/2020] [Indexed: 02/08/2023]
Abstract
The prevalence of obesity is rising every year and associated comorbidities such as cardiovascular diseases are among the leading causes of death worldwide. The gut microbiota has recently emerged as a potential target for therapeutic applications to prevent and treat those comorbidities. In this review, we focus on three conditions related to obesity in which the use of gut microbiota modulators could have benefits; mood disorders, eating behaviors, and body detoxification of persistent organic pollutants (POPs). On one hand, modulation of gut-derived signals to the brain in a context of obesity is involved in the development of neuroinflammation and can subsequently alter behaviors. An altered gut microbiome could change these signals and alleviate their consequences. On the other hand, obesity is associated with an increased accumulation of lipophilic contaminants, such as POPs. Targeting the microbiota could help body detoxication by reducing bioavailability, enhancing degradation by bioremediation or their excretion through the enterohepatic circulation. Thus, a supplementation of prebiotics, probiotics, or synbiotics could represent a complementary strategy to current ones, such as medication and lifestyle modifications, to decrease depression, alter eating behaviors, and lower body burden of pollutants considering the actual obesity epidemic our society is facing.
Collapse
|
44
|
Järbrink-Sehgal E, Andreasson A. The gut microbiota and mental health in adults. Curr Opin Neurobiol 2020; 62:102-114. [DOI: 10.1016/j.conb.2020.01.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 01/22/2020] [Accepted: 01/26/2020] [Indexed: 12/18/2022]
|
45
|
Longitudinal relationship of amino acids and indole metabolites with long-term body mass index and cardiometabolic risk markers in young individuals. Sci Rep 2020; 10:6399. [PMID: 32286421 PMCID: PMC7156759 DOI: 10.1038/s41598-020-63313-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/24/2020] [Indexed: 02/06/2023] Open
Abstract
Amino acid metabolites in biofluids are associated with high body mass index (BMI) and cardiometabolic abnormalities. However, prospective investigations regarding these associations are few, particularly among young individuals. Moreover, little is presently known about the impact of long-term high BMI. Using data from the DOrtmund Nutritional and Anthropometric Longitudinally Designed study (111 males and 107 females), we prospectively investigated relations between repeatedly measured urinary levels of 33 metabolites and (1) previously identified long-term BMI trajectory groups from childhood into late adolescence and (2) cardiometabolic risk markers in late adolescence–young adulthood, in sex-specific linear mixed regression models. Males with long-term overweight had lower indole-3-acetic acid when compared to others. Further, methionine, isoleucine, tryptophan, xanthurenic acid, and indole-3-carboxaldehyde were negatively associated with C-reactive protein (CRP), but 5-hydroxyindole-3-acetic acid was positively associated with CRP. No associations were observed in females. Long-term overweight from childhood into late adolescence is associated with decreased urinary levels of gut bacteria-derived indole-3-acetic acid, and several urinary amino acids, including gut bacteria-derived indole-3-carboxaldehyde are associated with elevated CRP later on in life. Taken together, our data suggest that indole metabolites, and their gut bacteria producers play potentially important roles in overweight-related inflammation.
Collapse
|
46
|
Maxwell AL, Gardiner E, Loxton NJ. Investigating the relationship between reward sensitivity, impulsivity, and food addiction: A systematic review. EUROPEAN EATING DISORDERS REVIEW 2020; 28:368-384. [PMID: 32142199 DOI: 10.1002/erv.2732] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/09/2020] [Accepted: 02/23/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The aim of the current research was to identify the extent to which reward sensitivity and impulsivity were related to food addiction. METHOD Forty-five studies, published from 2009 to June 2019, investigating reward sensitivity and/or impulsivity with food addiction as measured by the Yale Food Addiction Scale were reviewed. RESULTS Reward sensitivity, as measured by the Sensitivity to Reward (SR) scale, was positively associated with food addiction in two studies, but failed to yield consistent results in other studies when measured with the Behavioral Inhibition/Behavioral Activation Scales. Self-report impulsivity, as measured by the Barratt Impulsiveness Scale (BIS-11), was consistently associated with food addiction, with attentional impulsivity and motor impulsivity the most consistent subscales. Similarly, food addiction was also consistently associated with Negative Urgency, Positive Urgency, and Lack of Perseverance as measured by the UPPS-P Impulsive Behavior Scale. Food addiction was inconsistently associated with disinhibition, as measured by behavioral tasks, indicating food addiction appears more aligned with self-report measures of impulsivity. CONCLUSIONS Research in this field is dominated by university student, overweight and obese samples. Additional research is required to further tease out these relationships.
Collapse
Affiliation(s)
- Aimee L Maxwell
- School of Applied Psychology, Griffith University, Mt Gravatt Campus, Brisbane, Queensland, Australia
| | - Elliroma Gardiner
- School of Management, QUT Gardens Point, Brisbane, Queensland, Australia
| | - Natalie J Loxton
- School of Applied Psychology, Griffith University, Mt Gravatt Campus, Brisbane, Queensland, Australia.,Centre for Youth Substance Abuse Research, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
47
|
Sfera A, Osorio C, Diaz EL, Maguire G, Cummings M. The Other Obesity Epidemic-Of Drugs and Bugs. Front Endocrinol (Lausanne) 2020; 11:488. [PMID: 32849279 PMCID: PMC7411001 DOI: 10.3389/fendo.2020.00488] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 06/22/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic psychiatric patients with schizophrenia and related disorders are frequently treatment-resistant and may require higher doses of psychotropic drugs to remain stable. Prolonged exposure to these agents increases the risk of weight gain and cardiometabolic disorders, leading to poorer outcomes and higher medical cost. It is well-established that obesity has reached epidemic proportions throughout the world, however it is less known that its rates are two to three times higher in mentally ill patients compared to the general population. Psychotropic drugs have emerged as a major cause of weight gain, pointing to an urgent need for novel interventions to attenuate this unintended consequence. Recently, the gut microbial community has been linked to psychotropic drugs-induced obesity as these agents were found to possess antimicrobial properties and trigger intestinal dysbiosis, depleting Bacteroidetes phylum. Since germ-free animals exposed to psychotropics have not demonstrated weight gain, altered commensal flora composition is believed to be necessary and sufficient to induce dysmetabolism. Conversely, not only do psychotropics disrupt the composition of gut microbiota but the later alter the metabolism of the former. Here we review the role of gut bacterial community in psychotropic drugs metabolism and dysbiosis. We discuss potential biomarkers reflecting the status of Bacteroidetes phylum and take a closer look at nutritional interventions, fecal microbiota transplantation, and transcranial magnetic stimulation, strategies that may lower obesity rates in chronic psychiatric patients.
Collapse
Affiliation(s)
- Adonis Sfera
- Psychiatry, Loma Linda University, Loma Linda, CA, United States
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
- *Correspondence: Adonis Sfera
| | - Carolina Osorio
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
| | - Eddie Lee Diaz
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| | - Gerald Maguire
- Department of Psychiatry, University of California, Riverside, Riverside, CA, United States
| | - Michael Cummings
- Department of Psychiatry, Patton State Hospital, San Bernardino, CA, United States
| |
Collapse
|
48
|
Abstract
Preclinical evidence strongly suggests a role for the gut microbiome in modulating the host central nervous system function and behavior. Several communication channels have been identified that enable microbial signals to reach the brain and that enable the brain to influence gut microbial composition and function. In rodent models, endocrine, neural, and inflammatory signals generated by gut microbes can alter brain structure and function, while autonomic nervous system activity can affect the microbiome by modulating the intestinal environment and by directly regulating microbial behavior. The amount of information that reaches the brain is dynamically regulated by the blood-brain barrier and the intestinal barrier. In humans, associations between gut microbial composition and function and several brain disorders have been reported, and fecal microbial transplants from patient populations into gnotobiotic mice have resulted in the reproduction of homologous features in the recipient mice. However, in contrast to preclinical findings, there is little information about a causal role of the gut microbiome in modulating human central nervous system function and behavior. Longitudinal studies in large patient populations with therapeutic interventions are required to demonstrate such causality, which will provide the basis for future clinical trials. © 2020 American Physiological Society. Compr Physiol 10:57-72, 2020.
Collapse
Affiliation(s)
- Vadim Osadchiy
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, and UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Clair R Martin
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, and UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, UCLA Vatche and Tamar Manoukian Division of Digestive Diseases, and UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
49
|
Gheorghe CE, Martin JA, Manriquez FV, Dinan TG, Cryan JF, Clarke G. Focus on the essentials: tryptophan metabolism and the microbiome-gut-brain axis. Curr Opin Pharmacol 2019; 48:137-145. [PMID: 31610413 DOI: 10.1016/j.coph.2019.08.004] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 08/26/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
Abstract
The gut-brain axis is a bidirectional communication system between the central nervous system and the gastrointestinal tract, in which serotonin (5-HT) functions as a key neurotransmitter. Recent research has increasingly concentrated on tryptophan, the precursor to 5-HT and on the microbial regulation of tryptophan metabolism, with an emphasis on host-microbe control over kynurenine pathway metabolism and microbial-specific pathways that generate bioactive tryptophan metabolites. Here, we critically assess recent progress made towards a mechanistic understanding of the microbial regulation of tryptophan metabolism and microbiota-gut-brain axis homeostasis highlighting the role tryptophan metabolism plays in preclinical and clinical neuroscience and in the challenge to improve our understanding of how perturbed tryptophan metabolism contributes to stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Cassandra Elise Gheorghe
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jason A Martin
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Francisca Villalobos Manriquez
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland; INFANT Research Centre, University College Cork, Cork, Ireland.
| |
Collapse
|
50
|
Osadchiy V, Mayer EA, Bhatt R, Labus JS, Gao L, Kilpatrick LA, Liu C, Tillisch K, Naliboff B, Chang L, Gupta A. History of early life adversity is associated with increased food addiction and sex-specific alterations in reward network connectivity in obesity. Obes Sci Pract 2019; 5:416-436. [PMID: 31687167 PMCID: PMC6819979 DOI: 10.1002/osp4.362] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/17/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Neuroimaging studies have identified obesity-related differences in the brain's resting state activity. An imbalance between homeostatic and reward aspects of ingestive behaviour may contribute to obesity and food addiction. The interactions between early life adversity (ELA), the reward network and food addiction were investigated to identify obesity and sex-related differences, which may drive obesity and food addiction. METHODS Functional resting state magnetic resonance imaging was acquired in 186 participants (high body mass index [BMI]: ≥25: 53 women and 54 men; normal BMI: 18.50-24.99: 49 women and 30 men). Participants completed questionnaires to assess ELA (Early Traumatic Inventory) and food addiction (Yale Food Addiction Scale). A tripartite network analysis based on graph theory was used to investigate the interaction between ELA, brain connectivity and food addiction. Interactions were determined by computing Spearman rank correlations, thresholded at q < 0.05 corrected for multiple comparisons. RESULTS Participants with high BMI demonstrate an association between ELA and food addiction, with reward regions playing a role in this interaction. Among women with high BMI, increased ELA was associated with increased centrality of reward and emotion regulation regions. Men with high BMI showed associations between ELA and food addiction with somatosensory regions playing a role in this interaction. CONCLUSIONS The findings suggest that ELA may alter brain networks, leading to increased vulnerability for food addiction and obesity later in life. These alterations are sex specific and involve brain regions influenced by dopaminergic or serotonergic signalling.
Collapse
Affiliation(s)
- V. Osadchiy
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- David Geffen School of MedicineUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| | - E. A. Mayer
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- David Geffen School of MedicineUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- Vatche and Tamar Manoukin Division of Digestive DiseasesUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- Ahmanson‐Lovelace Brain Mapping CenterUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| | - R. Bhatt
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- Pediatric Pain and Palliative Care ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| | - J. S. Labus
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- David Geffen School of MedicineUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- Vatche and Tamar Manoukin Division of Digestive DiseasesUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| | - L. Gao
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| | - L. A. Kilpatrick
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- David Geffen School of MedicineUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- Vatche and Tamar Manoukin Division of Digestive DiseasesUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| | - C. Liu
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- David Geffen School of MedicineUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- Vatche and Tamar Manoukin Division of Digestive DiseasesUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| | - K. Tillisch
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- David Geffen School of MedicineUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- Vatche and Tamar Manoukin Division of Digestive DiseasesUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- Pediatric Pain and Palliative Care ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| | - B. Naliboff
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- David Geffen School of MedicineUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- Vatche and Tamar Manoukin Division of Digestive DiseasesUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| | - L. Chang
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- David Geffen School of MedicineUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- Vatche and Tamar Manoukin Division of Digestive DiseasesUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| | - A. Gupta
- G. Oppenheimer Family Center for Neurobiology of Stress and Resilience, Ingestive Behavior and Obesity ProgramUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- David Geffen School of MedicineUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
- Vatche and Tamar Manoukin Division of Digestive DiseasesUniversity of California, Los Angeles (UCLA)Los AngelesCAUSA
| |
Collapse
|