1
|
Crain E, Minaya DM, de La Serre CB. Microbiota-induced inflammation mediates the impacts of a Western diet on hippocampal-dependent memory. Nutr Res 2025; 138:89-106. [PMID: 40339190 DOI: 10.1016/j.nutres.2025.04.002] [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: 05/13/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 05/10/2025]
Abstract
Obesity is associated with impaired hippocampal-dependent memory, but the mechanisms driving this pathology are not fully understood. Western diets (WD) contribute to obesity, and previous reviews have described a role for WD in impaired hippocampal-dependent memory. However, there is need for a more detailed description of the pathways by which WD may impair memory. The short vs long-term effect of specific dietary components on brain structure and functions as well as the precise mechanism and molecular pathways involved are still not fully understood. This review focuses on the mechanisms and effects of gut microbiota-driven neuroinflammation. WD leads to changes and imbalance in bacterial taxa abundances that are deleterious to the host health (gut dysbiosis) and studies in rodent models show these changes are sufficient to impair hippocampal-dependent memory. Here, we discuss a variety of proposed mechanisms linking microbiota composition to hippocampal function, with a focus on neuroinflammation. Gut microbiota impacts gastrointestinal barrier function, leading to increased circulating proinflammatory bacterial products, increased blood-brain barrier permeability, and neuroinflammation.
Collapse
Affiliation(s)
- Eden Crain
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA
| | - Dulce M Minaya
- Department of Nutritional Sciences, University of Georgia, Athens, GA, USA
| | - Claire B de La Serre
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
2
|
Seong HJ, Park YM, Kim BS, Yoo HJ, Kim T, Yoon SM, Kim JH, Lee SY, Lee YK, Lee DW, Nam MH, Hong SJ. Integrated multi-omics reveals different host crosstalk of atopic dermatitis-enriched Bifidobacterium longum Strains. NPJ Biofilms Microbiomes 2025; 11:91. [PMID: 40442154 PMCID: PMC12122682 DOI: 10.1038/s41522-025-00714-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 04/25/2025] [Indexed: 06/02/2025] Open
Abstract
The infant gut microbiome is essential for long-term health and is linked to atopic dermatitis (AD), although the underlying mechanisms are not fully understood. This study investigated gut microbiome-host interactions in 31 infants with AD and 29 healthy controls using multi-omics approaches, including metagenomic, host transcriptomic, and metabolomic analyses. Microbial diversity was significantly altered in AD, with Bifidobacterium longum and Clostridium innocuum associated with these changes. At the strain-level, only B. longum differed significantly between groups, with pangenome analyses identifying genetic variations potentially affecting amino acid and lipid metabolites. Notably, B. longum subclade I, which was more prevalent in healthy controls, correlated with host transcriptomic pathways involved in phosphatidylinositol 3-kinase-AKT signaling and neuroactive ligand-receptor pathways, as well as specific metabolites, including tetrahydrocortisol and ornithine. These findings highlight the role of B. longum strain-level variation in infants, offering new insights into microbiome-host interactions related to AD.
Collapse
Grants
- This study was supported by the Bio & Medical Technology Development Program of the National Research Foundation of Korea (NRF), funded by the Korean government (MSIT) (NRF-2017M3A9F3043834) and the Korean Centers for Disease Control and Prevention (2008-E33030-00, 2009-E33033-00, 2011-E33021-00, 2012-E33012-00, 2013-E51003-00, 2014-E51004-00, 2014-E51004-01, 2014-E51004-02, 2017-E67002-00, 2017-E67002-01, 2017-E67002-02, 2020E670200, 2020E670201 and 2020E670202).
Collapse
Affiliation(s)
- Hoon Je Seong
- Department of Biological Science, Kunsan National University, Gunsan, Republic of Korea
| | - Yoon Mee Park
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Bong-Soo Kim
- Department of Life Science, Hallym University, Chuncheon, Republic of Korea
- Department of Nutritional Science and Food Management, Ewha Womans University, Seoul, Republic of Korea
| | - Hyun Ju Yoo
- Department of Convergence Medicine, Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Taeyune Kim
- Bioinformatics Institute, Macrogen, Inc., Seoul, Republic of Korea
| | - Sun Mi Yoon
- Meta Analysis Team, Macrogen, Inc., Seoul, Republic of Korea
| | - Jeong-Hyun Kim
- Department of Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - So-Yeon Lee
- PHI Digital Healthcare, Seoul, Republic of Korea
| | - Yun Kyung Lee
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bioscience, Soonchunhyang University, Cheonan, Republic of Korea
| | - Dong-Woo Lee
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Myung Hee Nam
- Seoul Center, Korea Basic Science Institute, Seoul, South Korea
| | - Soo-Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Li Q, Zhang Y, Wang X, Dai L, Zhao W. Gut microbiota of patients with post-stroke depression in Chinese population: a systematic review and meta-analysis. Front Cell Infect Microbiol 2025; 15:1444793. [PMID: 40375894 PMCID: PMC12078233 DOI: 10.3389/fcimb.2025.1444793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 03/17/2025] [Indexed: 05/18/2025] Open
Abstract
Background Evidence of changes in the composition and function of the gut microbiota (GM) in post-stroke depression (PSD) patients is gradually accumulating. This study aimed to systematically evaluate the relationship between PSD and GM. Methods We searched in PubMed, Web of Science, Embase, Cochrane databases, Wangfang, VIP, CBM, and CNKI from the establishment of the database to April 17, 2024, and systematic review and meta-analysis were performed to investigate the differences of GM between patients with PSD spectrum and healthy controls (HC) or stroke spectrum. Result There were 14 studies consisting a total of 1,556 individuals included in the meta-analysis. The pooled results showed that PSD spectrum demonstrated significantly increased α diversity as indexed by Chao1 index, ACE indexes, Shannon index, and Simpson index as compared to HC. Additionally, stroke spectrum significantly increased α diversity as indexed by Simpson index compared to PSD. Furthermore, the pooled estimation of relative abundance showed that Bacteroidota, Fusobacteriota, and Pseudomonadota in PSD patients were significantly higher than those in the HC group, while the abundance of Bacillota was higher in the HC group. Moreover, significant differences in GM were observed between PSD patients and HC at the family and genus levels. Conclusion This study found that the α diversity of PSD patients was higher than that of HC. Moreover, there were also differences in the distribution of GM at the phylum, family, and genus levels, respectively. At the same time, the level of Lachnospira in PSD patients was lower than that in the stroke group. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42024582708.
Collapse
Affiliation(s)
- Qiaoling Li
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Yuejuan Zhang
- Department of Nursing, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xiaoqian Wang
- Department of Nursing, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Lin Dai
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Wenli Zhao
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
4
|
Yassin LK, Nakhal MM, Alderei A, Almehairbi A, Mydeen AB, Akour A, Hamad MIK. Exploring the microbiota-gut-brain axis: impact on brain structure and function. Front Neuroanat 2025; 19:1504065. [PMID: 40012737 PMCID: PMC11860919 DOI: 10.3389/fnana.2025.1504065] [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] [Received: 09/30/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
The microbiota-gut-brain axis (MGBA) plays a significant role in the maintenance of brain structure and function. The MGBA serves as a conduit between the CNS and the ENS, facilitating communication between the emotional and cognitive centers of the brain via diverse pathways. In the initial stages of this review, we will examine the way how MGBA affects neurogenesis, neuronal dendritic morphology, axonal myelination, microglia structure, brain blood barrier (BBB) structure and permeability, and synaptic structure. Furthermore, we will review the potential mechanistic pathways of neuroplasticity through MGBA influence. The short-chain fatty acids (SCFAs) play a pivotal role in the MGBA, where they can modify the BBB. We will therefore discuss how SCFAs can influence microglia, neuronal, and astrocyte function, as well as their role in brain disorders such as Alzheimer's disease (AD), and Parkinson's disease (PD). Subsequently, we will examine the technical strategies employed to study MGBA interactions, including using germ-free (GF) animals, probiotics, fecal microbiota transplantation (FMT), and antibiotics-induced dysbiosis. Finally, we will examine how particular bacterial strains can affect brain structure and function. By gaining a deeper understanding of the MGBA, it may be possible to facilitate research into microbial-based pharmacological interventions and therapeutic strategies for neurological diseases.
Collapse
Affiliation(s)
- Lidya K. Yassin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammed M. Nakhal
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Alreem Alderei
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Afra Almehairbi
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ayishal B. Mydeen
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Amal Akour
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
5
|
Shang W, Huang Y, Xu Z, Li L, Gu Z, Cheng L, Hong Y. The impact of a high-carbohydrate diet on the cognitive behavior of mice in a low-pressure, low-oxygen environment. Food Funct 2025; 16:1116-1129. [PMID: 39831444 DOI: 10.1039/d4fo04831h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The effectiveness of high-carbohydrate diets (HCD) on cognitive impairment is still being debated. To clarify the impact of HCD on the cognitive behavior of mice under low-pressure hypoxic conditions, we studied 24 mice in different environments while subjecting them to dietary intervention for 5 weeks, and conducting behavioral tests. Under low-pressure hypoxic conditions, HCD intervention reversed the decline in spatial learning and memory abilities in mice caused by hypoxia, ameliorated pathological brain damage, and restored the integrity of the intestinal mucosa. We also identified differences in the microbial community. Under low-pressure hypoxic conditions, the intestinal abundance of Parasutterella in mice decreased, the abundance of harmful bacteria such as Desulfovibrio increased, and apoptosis was more prevalent, possibly explaining the observed decreases in glutathione peroxidase activity and brain-derived neurotrophic factor (BDNF) expression in the brain. HCD intervention increased the intestinal abundance of Bifidobacterium in hypoxic mice, reduced the abundances of Desulfovibrio and Faecalibaculum, and played antioxidant roles by lowering malondialdehyde levels and increasing superoxide dismutase activity in the brain by metabolizing amino acids and lipids. HCD also upregulated hippocampal BDNF levels and downregulated caspase 3. Collectively, these results are important because they help explain how HCD intervention can reduce hypoxia-induced damage to brain function.
Collapse
Affiliation(s)
- Weixuan Shang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yali Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhiqiang Xu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Lingjin Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhengbiao Gu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Li Cheng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yan Hong
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- Jiaxing Institute of Future Food, Jiaxing, 314000, China
| |
Collapse
|
6
|
Huang R, Liu Y. Efficacy of bifidobacterium-related preparations on depression: the first meta-analysis. Front Psychiatry 2024; 15:1463848. [PMID: 39421068 PMCID: PMC11484414 DOI: 10.3389/fpsyt.2024.1463848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/05/2024] [Indexed: 10/19/2024] Open
Abstract
Currently, depression-induced suicide has emerged as the primary contributor to the worldwide burden of disability. However, the prevailing drug treatment not only suffers from delayed effectiveness and limited efficacy, but also there are withdrawal symptoms and rebound phenomenon. Consequently, there is an imperative to investigate safer and more efficient treatments to ameliorate the clinical manifestations of depression. At present, there is increased evidence that probiotics can improve the symptoms of depression, but the existing studies use many and mixed types of probiotics, and it is impossible to determine the specific efficacy of bifidobacteria in the treatment of depression. This review will systematically review the effects of bifidobacteria on the treatment effect of depression, Meta-analysis showed that Bifidobacterium-related preparations effectively improved depressive symptoms in patients with depression. This study represents the initial meta-analysis conducted on the use of bifidobacteria-related agents for treating depression. The objective was to determine the effect of bifidobacteria-related preparations on improving depressive symptoms. We found that Bifidobacterium and its related agents can effectively reduce depression scale scores in patients with depression, suggesting the need for further research into this potential strategy for the prevention and treatment of depression.
Collapse
Affiliation(s)
- Ruihan Huang
- School of Public Health, Qilu Medical University, Zibo, Shandong, China
| | - Yongsheng Liu
- School of Clinical Medicine, Qilu Medical University, Zibo, Shandong, China
| |
Collapse
|
7
|
Zajkowska I, Niczyporuk P, Urbaniak A, Tomaszek N, Modzelewski S, Waszkiewicz N. Investigating the Impacts of Diet, Supplementation, Microbiota, Gut-Brain Axis on Schizophrenia: A Narrative Review. Nutrients 2024; 16:2228. [PMID: 39064675 PMCID: PMC11279812 DOI: 10.3390/nu16142228] [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: 06/24/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Schizophrenia is a disease with a complex etiology that significantly impairs the functioning of patients. In recent years, there has been increasing focus on the importance of the gut microbiota in the context of the gut-brain axis. In our study, we analyzed data on the gut-brain axis in relation to schizophrenia, as well as the impacts of eating habits, the use of various supplements, and diets on schizophrenia. Additionally, the study investigated the impact of antipsychotics on the development of metabolic disorders, such as diabetes, dyslipidemia, and obesity. There may be significant clinical benefits to be gained from therapies supported by supplements such as omega-3 fatty acids, B vitamins, and probiotics. The results suggest the need for a holistic approach to the treatment of schizophrenia, incorporating both drug therapy and dietary interventions.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Modzelewski
- Department of Psychiatry, Medical University of Bialystok, pl. Wołodyjowskiego 2, 15-272 Białystok, Poland; (I.Z.); (N.W.)
| | | |
Collapse
|
8
|
Roussin L, Gry E, Macaron M, Ribes S, Monnoye M, Douard V, Naudon L, Rabot S. Microbiota influence on behavior: Integrative analysis of serotonin metabolism and behavioral profile in germ-free mice. FASEB J 2024; 38:e23648. [PMID: 38822661 PMCID: PMC12086753 DOI: 10.1096/fj.202400334r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/05/2024] [Accepted: 04/22/2024] [Indexed: 06/03/2024]
Abstract
Previous studies on germ-free (GF) animals have described altered anxiety-like and social behaviors together with dysregulations in brain serotonin (5-HT) metabolism. Alterations in circulating 5-HT levels and gut 5-HT metabolism have also been reported in GF mice. In this study, we conducted an integrative analysis of various behaviors as well as markers of 5-HT metabolism in the brain and along the GI tract of GF male mice compared with conventional (CV) ones. We found a strong decrease in locomotor activity, accompanied by some signs of increased anxiety-like behavior in GF mice compared with CV mice. Brain gene expression analysis showed no differences in HTR1A and TPH2 genes. In the gut, we found decreased TPH1 expression in the colon of GF mice, while it was increased in the cecum. HTR1A expression was dramatically decreased in the colon, while HTR4 expression was increased both in the cecum and colon of GF mice compared with CV mice. Finally, SLC6A4 expression was increased in the ileum and colon of GF mice compared with CV mice. Our results add to the evidence that the microbiota is involved in regulation of behavior, although heterogeneity among studies suggests a strong impact of genetic and environmental factors on this microbiota-mediated regulation. While no impact of GF status on brain 5-HT was observed, substantial differences in gut 5-HT metabolism were noted, with tissue-dependent results indicating a varying role of microbiota along the GI tract.
Collapse
Affiliation(s)
- Léa Roussin
- Université Paris‐Saclay, INRAE, AgroParisTechMicalis InstituteJouy‐en‐JosasFrance
| | - Elisa Gry
- Université Paris‐Saclay, INRAE, AgroParisTechMicalis InstituteJouy‐en‐JosasFrance
| | - Mira Macaron
- Université Paris‐Saclay, INRAE, AgroParisTechMicalis InstituteJouy‐en‐JosasFrance
| | - Sandy Ribes
- Université Paris‐Saclay, INRAE, AgroParisTechMicalis InstituteJouy‐en‐JosasFrance
| | - Magali Monnoye
- Université Paris‐Saclay, INRAE, AgroParisTechMicalis InstituteJouy‐en‐JosasFrance
| | - Véronique Douard
- Université Paris‐Saclay, INRAE, AgroParisTechMicalis InstituteJouy‐en‐JosasFrance
| | - Laurent Naudon
- Université Paris‐Saclay, INRAE, AgroParisTech, CNRSMicalis InstituteJouy‐en‐JosasFrance
| | - Sylvie Rabot
- Université Paris‐Saclay, INRAE, AgroParisTechMicalis InstituteJouy‐en‐JosasFrance
| |
Collapse
|
9
|
Hung LY, Margolis KG. Autism spectrum disorders and the gastrointestinal tract: insights into mechanisms and clinical relevance. Nat Rev Gastroenterol Hepatol 2024; 21:142-163. [PMID: 38114585 DOI: 10.1038/s41575-023-00857-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 12/21/2023]
Abstract
Autism spectrum disorders (ASDs) are recognized as central neurodevelopmental disorders diagnosed by impairments in social interactions, communication and repetitive behaviours. The recognition of ASD as a central nervous system (CNS)-mediated neurobehavioural disorder has led most of the research in ASD to be focused on the CNS. However, gastrointestinal function is also likely to be affected owing to the neural mechanistic nature of ASD and the nervous system in the gastrointestinal tract (enteric nervous system). Thus, it is unsurprising that gastrointestinal disorders, particularly constipation, diarrhoea and abdominal pain, are highly comorbid in individuals with ASD. Gastrointestinal problems have also been repeatedly associated with increased severity of the core symptoms diagnostic of ASD and other centrally mediated comorbid conditions, including psychiatric issues, irritability, rigid-compulsive behaviours and aggression. Despite the high prevalence of gastrointestinal dysfunction in ASD and its associated behavioural comorbidities, the specific links between these two conditions have not been clearly delineated, and current data linking ASD to gastrointestinal dysfunction have not been extensively reviewed. This Review outlines the established and emerging clinical and preclinical evidence that emphasizes the gut as a novel mechanistic and potential therapeutic target for individuals with ASD.
Collapse
Affiliation(s)
- Lin Y Hung
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
| | - Kara Gross Margolis
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA.
- Department of Cell Biology, NYU Grossman School of Medicine and Langone Medical Center, New York, NY, USA.
- Department of Pediatrics, NYU Grossman School of Medicine and Langone Medical Center, New York, NY, USA.
| |
Collapse
|
10
|
Gan Y, Chen Y, Zhong H, Liu Z, Geng J, Wang H, Wang W. Gut microbes in central nervous system development and related disorders. Front Immunol 2024; 14:1288256. [PMID: 38343438 PMCID: PMC10854220 DOI: 10.3389/fimmu.2023.1288256] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/22/2023] [Indexed: 02/15/2024] Open
Abstract
The association between gut microbiota and central nervous system (CNS) development has garnered significant research attention in recent years. Evidence suggests bidirectional communication between the CNS and gut microbiota through the brain-gut axis. As a long and complex process, CNS development is highly susceptible to both endogenous and exogenous factors. The gut microbiota impacts the CNS by regulating neurogenesis, myelination, glial cell function, synaptic pruning, and blood-brain barrier permeability, with implication in various CNS disorders. This review outlines the relationship between gut microbiota and stages of CNS development (prenatal and postnatal), emphasizing the integral role of gut microbes. Furthermore, the review explores the implications of gut microbiota in neurodevelopmental disorders, such as autism spectrum disorder, Rett syndrome, and Angelman syndrome, offering insights into early detection, prompt intervention, and innovative treatments.
Collapse
Affiliation(s)
- Yumeng Gan
- Department of Infectious Disease and Hepatic Disease, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yao Chen
- Department of Infectious Disease and Hepatic Disease, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Huijie Zhong
- Department of Infectious Disease and Hepatic Disease, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Zhuo Liu
- Department of Infectious Disease and Hepatic Disease, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jiawei Geng
- Department of Infectious Disease and Hepatic Disease, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Huishan Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wenxue Wang
- Department of Infectious Disease and Hepatic Disease, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
- School of Basic Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
11
|
Filho AMC, Gomes NS, Lós DB, Leite IB, Tremblay MÈ, Macêdo DS. Microglia and Microbiome-Gut-Brain Axis. ADVANCES IN NEUROBIOLOGY 2024; 37:303-331. [PMID: 39207699 DOI: 10.1007/978-3-031-55529-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The mammalian gut contains a community of microorganisms called gut microbiome. The gut microbiome is integrated into mammalian physiology, contributing to metabolism, production of metabolites, and promoting immunomodulatory actions. Microglia, the brain's resident innate immune cells, play an essential role in homeostatic neurogenesis, synaptic remodeling, and glial maturation. Microglial dysfunction has been implicated in the pathogenesis of several neuropsychiatric disorders. Recent findings indicate that microglia are influenced by the gut microbiome and their derived metabolites throughout life. The pathways by which microbiota regulate microglia have only started to be understood, but this discovery has the potential to provide valuable insights into the pathogenesis of brain disorders associated with an altered microbiome. Here, we discuss the recent literature on the role of the gut microbiome in modulating microglia during development and adulthood and summarize the key findings on this bidirectional crosstalk in selected examples of neuropsychiatric and neurodegenerative disorders. We also highlight some current caveats and perspectives for the field.
Collapse
Affiliation(s)
- Adriano Maia Chaves Filho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Nayana Soares Gomes
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Deniele Bezerra Lós
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Isabel Bessa Leite
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Department of Molecular Medicine, Université de Laval, Québec City, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| | - Danielle S Macêdo
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
12
|
Moradian H, Gabriel T, Barrau M, Roblin X, Paul S. New methods to unveil host-microbe interaction mechanisms along the microbiota-gut-brain-axis. Gut Microbes 2024; 16:2351520. [PMID: 38717832 PMCID: PMC11086032 DOI: 10.1080/19490976.2024.2351520] [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: 10/20/2023] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
Links between the gut microbiota and human health have been supported throughout numerous studies, such as the development of neurological disease disorders. This link is referred to as the "microbiota-gut-brain axis" and is the focus of an emerging field of research. Microbial-derived metabolites and gut and neuro-immunological metabolites regulate this axis in health and many diseases. Indeed, assessing these signals, whether induced by microbial metabolites or neuro-immune mediators, could significantly increase our knowledge of the microbiota-gut-brain axis. However, this will require the development of appropriate techniques and potential models. Methods for studying the induced signals originating from the microbiota remain crucial in this field. This review discusses the methods and techniques available for studies of microbiota-gut-brain interactions. We highlight several much-debated elements of these methodologies, including the widely used in vivo and in vitro models, their implications, and perspectives in the field based on a systematic review of PubMed. Applications of various animal models (zebrafish, mouse, canine, rat, rabbit) to microbiota-gut-brain axis research with practical examples of in vitro methods and innovative approaches to studying gut-brain communications are highlighted. In particular, we extensively discuss the potential of "organ-on-a-chip" devices and their applications in this field. Overall, this review sheds light on the most widely used models and methods, guiding researchers in the rational choice of strategies for studies of microbiota-gut-brain interactions.
Collapse
Affiliation(s)
- Habibullah Moradian
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Saint-Etienne, France
| | - Tristan Gabriel
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Saint-Etienne, France
| | - Mathilde Barrau
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Xavier Roblin
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Stéphane Paul
- CIRI – Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, Saint-Etienne, France
- Immunology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
13
|
Cerdó T, Ruiz-Rodríguez A, Acuña I, Torres-Espínola FJ, Menchén-Márquez S, Gámiz F, Gallo M, Jehmlich N, Haange SB, von Bergen M, Campoy C, Suárez A. Infant gut microbiota contributes to cognitive performance in mice. Cell Host Microbe 2023; 31:1974-1988.e4. [PMID: 38052208 DOI: 10.1016/j.chom.2023.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/26/2023] [Accepted: 11/07/2023] [Indexed: 12/07/2023]
Abstract
Gut microbiota has been linked to infant neurodevelopment. Here, an association between infant composite cognition and gut microbiota composition is established as soon as 6 months. Higher diversity and evenness characterize microbial communities of infants with composite cognition above (Inf-aboveCC) versus below (Inf-belowCC) median values. Metaproteomic and metabolomic analyses establish an association between microbial histidine ammonia lyase and infant histidine metabolome with cognition. Fecal transplantation from Inf-aboveCC versus Inf-belowCC donors into germ-free mice shows that memory, assessed by a novel object recognition test, is a transmissible trait. Furthermore, Inf-aboveCC mice are enriched in species belonging to Phocaeicola, as well as Bacteroides and Bifidobacterium, previously linked to cognition. Finally, Inf-aboveCC mice show lower fecal histidine and urocanate:histidine and urocanate:glutamate ratios in the perirhinal cortex compared to Inf-belowCC mice. Overall, these findings reveal a causative role of gut microbiota on infant cognition, pointing at the modulation of histidine metabolite levels as a potential underlying mechanism.
Collapse
Affiliation(s)
- Tomás Cerdó
- Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Reina Sofia University Hospital, University of Córdoba, 14004 Córdoba, Spain; Centre for Rheumatology Research, Division of Medicine, University College London, London WC1E 6JF, UK
| | - Alicia Ruiz-Rodríguez
- Department of Biochemistry and Molecular Biology 2, Nutrition and Food Technology Institute "José Mataix" (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18016 Granada, Spain; Department of Microbiology, Nutrition and Food Technology Institute "José Mataix" (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18016 Granada, Spain.
| | - Inmaculada Acuña
- Department of Biochemistry and Molecular Biology 2, Nutrition and Food Technology Institute "José Mataix" (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18016 Granada, Spain
| | - Francisco José Torres-Espínola
- EURISTIKOS Excellence Centre for Pediatric Research, Institute of Neurosciences, Biomedical Research Centre (CIBM), University of Granada, 18016 Granada, Spain
| | - Sergio Menchén-Márquez
- Department of Psychobiology, Institute of Neurosciences, Biomedical Research Centre (CIBM), University of Granada, 18016 Granada, Spain
| | - Fernando Gámiz
- Department of Psychobiology, Institute of Neurosciences, Biomedical Research Centre (CIBM), University of Granada, 18016 Granada, Spain
| | - Milagros Gallo
- Department of Psychobiology, Institute of Neurosciences, Biomedical Research Centre (CIBM), University of Granada, 18016 Granada, Spain; Instituto de Investigación Biosanitaria (IBS), 18014 Granada, Spain
| | - Nico Jehmlich
- Department of Molecular System Biology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| | - Sven-Bastiaan Haange
- Department of Molecular System Biology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular System Biology, Helmholtz Centre for Environmental Research-UFZ, Permoserstraße 15, 04318 Leipzig, Germany
| | - Cristina Campoy
- EURISTIKOS Excellence Centre for Pediatric Research, Institute of Neurosciences, Biomedical Research Centre (CIBM), University of Granada, 18016 Granada, Spain; Instituto de Investigación Biosanitaria (IBS), 18014 Granada, Spain; Department of Pediatrics, School of Medicine, University of Granada, 18016 Granada, Spain
| | - Antonio Suárez
- Department of Biochemistry and Molecular Biology 2, Nutrition and Food Technology Institute "José Mataix" (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18016 Granada, Spain
| |
Collapse
|
14
|
Wang J, Qiu F, Zhang Z, Liu Y, Zhou Q, Dai S, Xiang S, Wei C. Clostridium butyricum Alleviates DEHP Plasticizer-Induced Learning and Memory Impairment in Mice via Gut-Brain Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:18524-18537. [PMID: 37963287 DOI: 10.1021/acs.jafc.3c03533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) plasticizer, a well-known environmental and food pollutant, has neurotoxicity. However, it is unknown whether DEHP leads to learning and memory impairment through gut-brain axis and whether Clostridium butyricum can alleviate this impairment. Here, C57BL/6 mice were exposed to DEHP and treated with C. butyricum. Learning and memory abilities were evaluated through the Morris water maze. The levels of synaptic proteins, inflammatory cytokines, and 5-hydroxytryptamine (5-HT) were detected by immunohistochemistry or ELISA. Gut microbiota were analyzed through 16S rRNA sequencing. C. butyricum alleviated DEHP-induced learning and memory impairment and restored synaptic proteins. It significantly relieved DEHP-induced inflammation and recovered 5-HT levels. C. butyricum recovered the richness of the gut microbiota decreased by DEHP, with the Bifidobacterium genus increasing the most. Overall, C. butyricum alleviated DEHP-induced learning and memory impairment due to reduced inflammation and increased 5-HT secretion, which was partly attributed to the recovery of gut microbiota.
Collapse
Affiliation(s)
- Jin Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Feng Qiu
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Zilong Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Yu Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Qian Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Siyu Dai
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Shuanglin Xiang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| | - Chenxi Wei
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha 410081, Hunan, China
| |
Collapse
|
15
|
Hinks M, Martin YH, Burke FF, Bambico FR, Swift-Gallant A. Cross-sex cecal microbiota transfer alters depressive-like behaviours in mice. Behav Processes 2023; 213:104960. [PMID: 37884083 DOI: 10.1016/j.beproc.2023.104960] [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: 06/16/2023] [Revised: 09/06/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023]
Abstract
Major depressive disorder (MDD) is a leading cause of non-fatal global disease burden, with females being two-fold more likely than males to be diagnosed with the disorder. Despite this sex-linked disparity of diagnosis, it is unclear what underlies the sex bias in MDD. Recent findings suggest a role for the gut in mediating affective disorders through the gut-brain-axis (GBA). However, few studies have included sex as a biological variable. For this study, cross-sex transfer of cecal microbiota was performed between male and female C57Bl/6 mice to elucidate the effects of sex and the gut microbiome on a standard battery of tests measuring depressive-like behaviours. Specifically, regardless of sex, recipients of male cecal content had a greater sucrose preference than controls and recipients of female cecum, although recipients of male cecal transfer also showed a trend towards increased passive coping on the force swim test. Conversely, in the splash test, recipients of female cecum displayed a decrease in grooming behaviour compared to both controls and recipients of male cecum, suggestive of an increase in depressive-like behaviour. These results support a role for female-specific gut microbes in contributing to female vulnerability to depression, particularly on self-care measures, while male-specific gut microbes may protect in part against an anhedonia-like phenotype, but increase passive coping.
Collapse
Affiliation(s)
- Meagan Hinks
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's, NL A1C 5S7, Canada
| | - Yellow H Martin
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's, NL A1C 5S7, Canada
| | - Francine F Burke
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's, NL A1C 5S7, Canada
| | - Francis R Bambico
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's, NL A1C 5S7, Canada
| | - Ashlyn Swift-Gallant
- Department of Psychology, Memorial University of Newfoundland and Labrador, St. John's, NL A1C 5S7, Canada.
| |
Collapse
|
16
|
Gutierrez A, Pucket B, Engevik MA. Bifidobacterium and the intestinal mucus layer. MICROBIOME RESEARCH REPORTS 2023; 2:36. [PMID: 38045921 PMCID: PMC10688832 DOI: 10.20517/mrr.2023.37] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/21/2023] [Accepted: 09/13/2023] [Indexed: 12/05/2023]
Abstract
Bifidobacterium species are integral members of the human gut microbiota and these microbes have significant interactions with the intestinal mucus layer. This review delves into Bifidobacterium-mucus dynamics, shedding light on the multifaceted nature of this relationship. We cover conserved features of Bifidobacterium-mucus interactions, such as mucus adhesion and positive regulation of goblet cell and mucus production, as well as species and strain-specific attributes of mucus degradation. For each interface, we explore the molecular mechanisms underlying these interactions and their potential implications for human health. Notably, we emphasize the ability of Bifidobacterium species to positively influence the mucus layer, shedding light on its potential as a mucin-builder and a therapeutic agent for diseases associated with disrupted mucus barriers. By elucidating the complex interplay between Bifidobacterium and intestinal mucus, we aim to contribute to a deeper understanding of the gut microbiota-host interface and pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Alyssa Gutierrez
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Brenton Pucket
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Melinda A. Engevik
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
17
|
Torices S, Daire L, Simon S, Mendoza L, Daniels D, Joseph JA, Fattakhov N, Naranjo O, Teglas T, Toborek M. The NLRP3 inflammasome and gut dysbiosis as a putative link between HIV-1 infection and ischemic stroke. Trends Neurosci 2023; 46:682-693. [PMID: 37330380 PMCID: PMC10554647 DOI: 10.1016/j.tins.2023.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/26/2023] [Accepted: 05/18/2023] [Indexed: 06/19/2023]
Abstract
HIV-associated comorbidities, such as ischemic stroke, are prevalent in people with HIV (PWH). Several studies both in animal models and humans have revealed an association between activation of the inflammasome in HIV-1 infection and stroke. The gut microbiota is an important component in controlling neuroinflammation in the CNS. It has also been proposed to be involved in the pathobiology of HIV-1 infection, and has been associated with an increase in activation of the inflammasome. In this review, we provide an overview of the microbiota-gut-inflammasome-brain axis, focusing on the NLRP3 inflammasome and dysregulation of the microbiome as risk factors that may contribute to the outcome of ischemic stroke and recovery in PWH. We also focus on the potential of targeting the NLRP3 inflammasome as a novel therapeutic approach for PWH who are at risk of developing cerebrovascular diseases.
Collapse
Affiliation(s)
- Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA.
| | - Leah Daire
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Sierra Simon
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Luisa Mendoza
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Destiny Daniels
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Joelle-Ann Joseph
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Nikolai Fattakhov
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Timea Teglas
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Gautier Bldg., Suite 528, 1011 NW 15th Street, Miami, FL 33136, USA.
| |
Collapse
|
18
|
Gancz NN, Levinson JA, Callaghan BL. Sex and gender as critical and distinct contributors to the human brain-gut-microbiome axis. Brain Res Bull 2023; 199:110665. [PMID: 37192716 PMCID: PMC11149430 DOI: 10.1016/j.brainresbull.2023.110665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/25/2023] [Accepted: 05/13/2023] [Indexed: 05/18/2023]
Abstract
The brain-gut-microbiome axis (BGMA) is a pivotal contributor to human health. A large body of research, especially from animal models, has revealed bidirectional, causal relationships between the BGMA and sex. In particular, sex steroids appear to be affected by the BGMA, to influence the BGMA, and to moderate environmental effects on the BGMA. However, animal research on the relationship between sex and the BGMA has not translated well to human models. We contend that this is due in part to an oversimplified approach to sex: although BGMA researchers have traditionally approached sex as a unidimensional, dichotomous variable, it is in fact multidimensional and is comprised of both multi-categorical and continuous dimensions. We also contend that research on the BGMA in humans should approach gender as a variable that is distinct from sex and that gender may influence the BGMA through pathways that are independent from the effects of sex alone. Research practices that consider the complexity and distinctiveness of sex and gender in relation to the human BGMA will not only yield improved understanding of this consequential system, but will also enhance the development of treatments for adverse health outcomes with BGMA-related etiologies. We conclude with recommendations for the implementation of such practices.
Collapse
Affiliation(s)
- N N Gancz
- University of California, Los Angeles, Department of Psychology, 502 Portola Plaza, Los Angeles, CA 90095, USA.
| | - J A Levinson
- University of California, Los Angeles, Department of Psychology, 502 Portola Plaza, Los Angeles, CA 90095, USA
| | - B L Callaghan
- University of California, Los Angeles, Department of Psychology, 502 Portola Plaza, Los Angeles, CA 90095, USA
| |
Collapse
|
19
|
Li J, Li D, Chen Y, Chen W, Xu J, Gao L. Gut Microbiota and Aging: Traditional Chinese Medicine and Modern Medicine. Clin Interv Aging 2023; 18:963-986. [PMID: 37351381 PMCID: PMC10284159 DOI: 10.2147/cia.s414714] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/08/2023] [Indexed: 06/24/2023] Open
Abstract
The changing composition of gut microbiota, much like aging, accompanies people throughout their lives, and the inextricable relationship between both has recently attracted extensive attention as well. Modern medical research has revealed that a series of changes in gut microbiota are involved in the aging process of organisms, which may be because gut microbiota modulates aging-related changes related to innate immunity and cognitive function. At present, there is no definite and effective method to delay aging. However, Nobel laureate Tu Youyou's research on artemisinin has inspired researchers to study the importance of Traditional Chinese Medicine (TCM). TCM, as an ancient alternative medicine, has unique advantages in preventive health care and in treating diseases as it already has formed an independent understanding of the aging system. TCM practitioners believe that the mechanism of aging is mainly deficiency, and pathological states such as blood stasis, qi stagnation and phlegm coagulation can exacerbate the process of aging, which involves a series of organs, including the brain, kidney, heart, liver and spleen. Our current understanding of aging has led us to realise that TCM can indeed make some beneficial changes, such as the improvement of cognitive impairment. However, due to the multi-component and multi-target nature of TCM, the exploration of its mechanism of action has become extremely complex. While analysing the relationship between gut microbiota and aging, this review explores the similarities and differences in treatment methods and mechanisms between TCM and Modern Medicine, in order to explore a new approach that combines TCM and Modern Medicine to regulate gut microbiota, improve immunity and delay aging.
Collapse
Affiliation(s)
- Jinfan Li
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, 250000, People’s Republic of China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Dong Li
- Department of Diabetes, Licheng District Hospital of Traditional Chinese Medicine, Jinan, Shandong, 250100, People’s Republic of China
| | - Yajie Chen
- Department of Rehabilitation and Health Care, Jinan Vocational College of Nursing, Jinan, Shandong, 250100, People’s Republic of China
| | - Wenbin Chen
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, People’s Republic of China
| | - Jin Xu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, People’s Republic of China
| | - Ling Gao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, 250021, People’s Republic of China
| |
Collapse
|
20
|
McMath AL, Aguilar-Lopez M, Cannavale CN, Khan NA, Donovan SM. A systematic review on the impact of gastrointestinal microbiota composition and function on cognition in healthy infants and children. Front Neurosci 2023; 17:1171970. [PMID: 37389363 PMCID: PMC10306408 DOI: 10.3389/fnins.2023.1171970] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/23/2023] [Indexed: 07/01/2023] Open
Abstract
Evidence from animal models or children with neurodevelopmental disorders has implicated the gut microbiome (GM) in neurocognitive development. However, even subclinical impairement of cognition can have negative consequences, as cognition serves as the foundation for skills necessary to succeed in school, vocation and socially. The present study aims to identify gut microbiome characteristics or changes in gut microbiome characteristics that consistently associate with cognitive outcomes in healthy, neurotypical infants and children. Of the 1,520 articles identified in the search, 23 were included in qualitative synthesis after applying exclusion criteria. Most studies were cross-sectional and focused on behavior or motor and language skills. Bifidobacterium, Bacteroides, Clostridia, Prevotella, and Roseburia were related to these aspects of cognition across several studies. While these results support the role of GM in cognitive development, higher quality studies focused on more complex cognition are needed to understand the extent to which the GM contributes to cognitive development.
Collapse
Affiliation(s)
- Arden L. McMath
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Miriam Aguilar-Lopez
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Texas Children’s Microbiome Center, Department of Pathology, Texas Children’s Hospital, Houston, TX, United States
| | - Corinne N. Cannavale
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Naiman A. Khan
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, United States
- Neuroscience Program, University of Illinois Urbana-Champaign, Champaign, IL, United States
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Champaign, IL, United States
| | - Sharon M. Donovan
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, United States
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
21
|
Kim CS, Shin DM. Gut microbiota and cognitive development in infant mice: Quantity and source of potable water. PLoS One 2023; 18:e0286951. [PMID: 37315057 DOI: 10.1371/journal.pone.0286951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/29/2023] [Indexed: 06/16/2023] Open
Abstract
Not only the water quantity consumed but also the source of drinking water has been considered for their health benefits, but there is limited evidence. We aimed to determine whether the amount and type of drinking water affect physiological and biological functions, including brain function, by confirming how it affects gut microbiota which has an important regulatory role in host physiology. Three-week-old infant mice were subjected to 1) a water restriction experiment (control group, ad libitum consumption of distilled water; dehydration group, time-limited access to distilled water [15 min/day]) and 2) different water source experiment (distilled water, purified water, spring water, and tap water groups). The gut microbiota and cognitive development were analyzed using the 16S ribosomal ribonucleic acid sequencing method and the Barnes maze, respectively. The relative abundance of Firmicutes and Bacteroidetes and the Firmicutes-to-Bacteroidetes ratio (F/B ratio) changed depending on age (juveniles vs. infants). Insufficient water intake reversed these developmental changes, showing that the relative abundances of Bacteroidetes and Firmicutes and the F/B ratio in dehydrated juvenile mice were similar to those in normal infant mice. Additionally, clustering analysis revealed no significant differences in the intestinal flora in the mice from the different drinking water sources; however, dehydration significantly altered the composition of the genera compared to the other water source groups wherein water was provided ad libitum. Moreover, cognitive development was significantly disrupted by insufficient water intake, although the type of drinking water had no significant influence. Cognitive decline, measured by relative latency, was positively associated with the relative abundance of unclassified Erysipelotrichaceae that were in significantly high relative abundance in the dehydration group. These results suggest that the water quantity consumed, rather than the mineral content of drinking water, is imperative for shaping the early gut microbiota associated with cognitive development during infancy.
Collapse
Affiliation(s)
- Chong-Su Kim
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul, Republic of Korea
- Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul, Republic of Korea
- Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
22
|
Interrogation of the mammalian gut-brain axis using LC-MS/MS-based targeted metabolomics with in vitro bacterial and organoid cultures and in vivo gnotobiotic mouse models. Nat Protoc 2023; 18:490-529. [PMID: 36352124 DOI: 10.1038/s41596-022-00767-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 07/26/2022] [Indexed: 11/11/2022]
Abstract
Interest in the communication between the gastrointestinal tract and central nervous system, known as the gut-brain axis, has prompted the development of quantitative analytical platforms to analyze microbe- and host-derived signals. This protocol enables investigations into connections between microbial colonization and intestinal and brain neurotransmitters and contains strategies for the comprehensive evaluation of metabolites in in vitro (organoids) and in vivo mouse model systems. Here we present an optimized workflow that includes procedures for preparing these gut-brain axis model systems: (stage 1) growth of microbes in defined media; (stage 2) microinjection of intestinal organoids; and (stage 3) generation of animal models including germ-free (no microbes), specific-pathogen-free (complete gut microbiota) and specific-pathogen-free re-conventionalized (germ-free mice associated with a complete gut microbiota from a specific-pathogen-free mouse), and Bifidobacterium dentium and Bacteroides ovatus mono-associated mice (germ-free mice colonized with a single gut microbe). We describe targeted liquid chromatography-tandem mass spectrometry-based metabolomics methods for analyzing microbially derived short-chain fatty acids and neurotransmitters from these samples. Unlike other protocols that commonly examine only stool samples, this protocol includes bacterial cultures, organoid cultures and in vivo samples, in addition to monitoring the metabolite content of stool samples. The incorporation of three experimental models (microbes, organoids and animals) enhances the impact of this protocol. The protocol requires 3 weeks of murine colonization with microbes and ~1-2 weeks for liquid chromatography-tandem mass spectrometry-based instrumental and quantitative analysis, and sample post-processing and normalization.
Collapse
|
23
|
Belyaeva IA, Bombardirova EP, Turti TV. New Strategies for Enhancement of Infant Milk Formulas Composition. CURRENT PEDIATRICS 2022. [DOI: 10.15690/vsp.v21i6.2468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This article covers the issues of remodeling breast milk’s protective properties during creating infant milk formulas. First of all, this concerns the conditions for normal intestinal microbiota development in growing organism. Its quantitative and qualitative features are the trigger of either sanogenetic, or pathological immune and metabolic reactions, and also determine gut-brain axis functioning. The protective significance of prebiotic composition diversity of mammalian milk and the inductive role of breast milk oligosaccharides are shown. The modern concept of synbiotics role in gastrointestinal tract and other systems functioning, as well as the use of modern synbiotics in the creation of infant formulas (available Russian formula included) are presented.
Collapse
Affiliation(s)
- I. A. Belyaeva
- Research Institute of Pediatrics and Children’s Health in Petrovsky National Research Centre of Surgery; Pirogov Russian National Research Medical University; Morozovskaya Children’s City Hospital
| | - E. P. Bombardirova
- Research Institute of Pediatrics and Children’s Health in Petrovsky National Research Centre of Surgery
| | - T. V. Turti
- Research Institute of Pediatrics and Children’s Health in Petrovsky National Research Centre of Surgery; Pirogov Russian National Research Medical University; Research Institute for Healthcare Organization and Medical Management
| |
Collapse
|
24
|
Vatanen T, Jabbar KS, Ruohtula T, Honkanen J, Avila-Pacheco J, Siljander H, Stražar M, Oikarinen S, Hyöty H, Ilonen J, Mitchell CM, Yassour M, Virtanen SM, Clish CB, Plichta DR, Vlamakis H, Knip M, Xavier RJ. Mobile genetic elements from the maternal microbiome shape infant gut microbial assembly and metabolism. Cell 2022; 185:4921-4936.e15. [PMID: 36563663 PMCID: PMC9869402 DOI: 10.1016/j.cell.2022.11.023] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 09/30/2022] [Accepted: 11/11/2022] [Indexed: 12/24/2022]
Abstract
The perinatal period represents a critical window for cognitive and immune system development, promoted by maternal and infant gut microbiomes and their metabolites. Here, we tracked the co-development of microbiomes and metabolomes from late pregnancy to 1 year of age using longitudinal multi-omics data from a cohort of 70 mother-infant dyads. We discovered large-scale mother-to-infant interspecies transfer of mobile genetic elements, frequently involving genes associated with diet-related adaptations. Infant gut metabolomes were less diverse than maternal but featured hundreds of unique metabolites and microbe-metabolite associations not detected in mothers. Metabolomes and serum cytokine signatures of infants who received regular-but not extensively hydrolyzed-formula were distinct from those of exclusively breastfed infants. Taken together, our integrative analysis expands the concept of vertical transmission of the gut microbiome and provides original insights into the development of maternal and infant microbiomes and metabolomes during late pregnancy and early life.
Collapse
Affiliation(s)
- Tommi Vatanen
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Terhi Ruohtula
- New Children's Hospital, Helsinki University Hospital, Helsinki, Finland
| | - Jarno Honkanen
- New Children's Hospital, Helsinki University Hospital, Helsinki, Finland
| | | | - Heli Siljander
- New Children's Hospital, Helsinki University Hospital, Helsinki, Finland; Centre for Military Medicine, Finnish Defence Forces, Riihimäki, Finland
| | - Martin Stražar
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sami Oikarinen
- Department of Virology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Heikki Hyöty
- Department of Virology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Fimlab Laboratories, Tampere, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Caroline M Mitchell
- Vincent Obstetrics & Gynecology Department, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Moran Yassour
- Microbiology & Molecular Genetics Department, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Suvi M Virtanen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland; Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland; Center for Child Health Research and Development and Innovation Center, Tampere University Hospital, Tampere, Finland
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Damian R Plichta
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Microbiome Informatics and Therapeutics, MIT, Cambridge, MA 02139, USA
| | - Hera Vlamakis
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Microbiome Informatics and Therapeutics, MIT, Cambridge, MA 02139, USA
| | - Mikael Knip
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Helsinki University Hospital, Helsinki, Finland; Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Microbiome Informatics and Therapeutics, MIT, Cambridge, MA 02139, USA; Center for Computational and Integrative Biology, Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
25
|
Lim S, Lee S. Chemical Modulators for Targeting Autism Spectrum Disorders: From Bench to Clinic. Molecules 2022; 27:molecules27165088. [PMID: 36014340 PMCID: PMC9414776 DOI: 10.3390/molecules27165088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Autism spectrum disorders (ASD) are neurodevelopmental disorders characterized by diverse behavioral symptoms such as repetitive behaviors, social deficits, anxiety, hyperactivity, and irritability. Despite their increasing incidence, the specific pathological mechanisms of ASD are still unknown, and the degree and types of symptoms that vary from patient to patient make it difficult to develop drugs that target the core symptoms of ASD. Although various atypical antipsychotics and antidepressants have been applied to regulate ASD symptoms, these drugs can only alleviate the symptoms and do not target the major causes. Therefore, development of novel drugs targeting factors directly related to the onset of ASD is required. Among the various factors related to the onset of ASD, several chemical modulators to treat ASD, focused on serotonin (5-hydroxytryptamine, 5-HT) and glutamate receptors, microbial metabolites, and inflammatory cytokines, are explored in this study. In particular, we focus on the chemical drugs that have improved various aspects of ASD symptoms in animal models and in clinical trials for various ages of patients with ASD.
Collapse
Affiliation(s)
- Songhyun Lim
- Creative Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Sanghee Lee
- Creative Research Center for Brain Science, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
- Department of HY-KIST Bio-Convergence, Hanyang University, Seoul 04763, Korea
- Correspondence: ; Tel.: +82-2-958-5138
| |
Collapse
|
26
|
Doroftei B, Ilie OD, Diaconu R, Hutanu D, Stoian I, Ilea C. An Updated Narrative Mini-Review on the Microbiota Changes in Antenatal and Post-Partum Depression. Diagnostics (Basel) 2022; 12:diagnostics12071576. [PMID: 35885482 PMCID: PMC9315700 DOI: 10.3390/diagnostics12071576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 12/02/2022] Open
Abstract
Background: Antenatal depression (AND) and post-partum depression (PPD) are long-term debilitating psychiatric disorders that significantly influence the composition of the gut flora of mothers and infants that starts from the intrauterine life. Not only does bacterial ratio shift impact the immune system, but it also increases the risk of potentially life-threatening disorders. Material and Methods: Therefore, we conducted a narrative mini-review aiming to gather all evidence published between 2018–2022 regarding microflora changes in all three stages of pregnancy. Results: We initially identified 47 potentially eligible studies, from which only 7 strictly report translocations; 3 were conducted on rodent models and 4 on human patients. The remaining studies were divided based on their topic, precisely focused on how probiotics, breastfeeding, diet, antidepressants, exogenous stressors, and plant-derived compounds modulate in a bidirectional way upon behavior and microbiota. Almost imperatively, dysbacteriosis cause cognitive impairments, reflected by abnormal temperament and personality traits that last up until 2 years old. Thankfully, a distinct technique that involves fecal matter transfer between individuals has been perfected over the years and was successfully translated into clinical practice. It proved to be a reliable approach in diminishing functional non- and gastrointestinal deficiencies, but a clear link between depressive women’s gastrointestinal/vaginal microbiota and clinical outcomes following reproductive procedures is yet to be established. Another gut-dysbiosis-driving factor is antibiotics, known for their potential to trigger inflammation. Fortunately, the studies conducted on mice that lack microbiota offer, without a shadow of a doubt, insight. Conclusions: It can be concluded that the microbiota is a powerful organ, and its optimum functionality is crucial, likely being the missing puzzle piece in the etiopathogenesis of psychiatric disorders.
Collapse
Affiliation(s)
- Bogdan Doroftei
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, No. 16, 700115 Iasi, Romania; (B.D.); (I.S.); (C.I.)
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street, No. 34, 700038 Iasi, Romania;
- Origyn Fertility Center, Palace Street, No. 3C, 700032 Iasi, Romania
| | - Ovidiu-Dumitru Ilie
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University, Carol I Avenue, No. 20A, 700505 Iasi, Romania
- Correspondence:
| | - Roxana Diaconu
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street, No. 34, 700038 Iasi, Romania;
- Origyn Fertility Center, Palace Street, No. 3C, 700032 Iasi, Romania
| | - Delia Hutanu
- Department of Biology, Faculty of Chemistry-Biology-Geography, West University of Timisoara, Vasile Pârvan Avenue, No. 4, 300115 Timisoara, Romania;
| | - Irina Stoian
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, No. 16, 700115 Iasi, Romania; (B.D.); (I.S.); (C.I.)
| | - Ciprian Ilea
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street, No. 16, 700115 Iasi, Romania; (B.D.); (I.S.); (C.I.)
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street, No. 34, 700038 Iasi, Romania;
| |
Collapse
|
27
|
Horvath TD, Ihekweazu FD, Haidacher SJ, Ruan W, Engevik KA, Fultz R, Hoch KM, Luna RA, Oezguen N, Spinler JK, Haag AM, Versalovic J, Engevik MA. Bacteroides ovatus colonization influences the abundance of intestinal short chain fatty acids and neurotransmitters. iScience 2022; 25:104158. [PMID: 35494230 PMCID: PMC9038548 DOI: 10.1016/j.isci.2022.104158] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 11/01/2021] [Accepted: 03/23/2022] [Indexed: 12/18/2022] Open
Abstract
Gut microbes can synthesize multiple neuro-active metabolites. We profiled neuro-active compounds produced by the gut commensal Bacteroides ovatus in vitro and in vivo by LC-MS/MS. We found that B. ovatus generates acetic acid, propionic acid, isobutyric acid, and isovaleric acid. In vitro, B. ovatus consumed tryptophan and glutamate and synthesized the neuro-active compounds glutamine and GABA. Consistent with our LC-MS/MS-based in vitro data, we observed elevated levels of acetic acid, propionic acid, isobutyric acid, and isovaleric acid in the intestines of B. ovatus mono-associated mice compared with germ-free controls. B. ovatus mono-association also increased the concentrations of intestinal GABA and decreased the concentrations of tryptophan and glutamine compared with germ-free controls. Computational network analysis revealed unique links between SCFAs, neuro-active compounds, and colonization status. These results highlight connections between microbial colonization and intestinal neurotransmitter concentrations, suggesting that B. ovatus selectively influences the presence of intestinal neurotransmitters.
Collapse
Affiliation(s)
- Thomas D. Horvath
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Faith D. Ihekweazu
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Section of Gastroenterology, Hepatology, and Nutrition, Texas Children’s Hospital, Houston, TX, USA
| | - Sigmund J. Haidacher
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Wenly Ruan
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Section of Gastroenterology, Hepatology, and Nutrition, Texas Children’s Hospital, Houston, TX, USA
| | - Kristen A. Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Robert Fultz
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, TX, USA
| | - Kathleen M. Hoch
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Ruth Ann Luna
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Numan Oezguen
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Jennifer K. Spinler
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Anthony M. Haag
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children’s Hospital, Houston, TX, USA
| | - Melinda A. Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, 173 Ashley Ave, BSB 621, Charleston, SC 29425, USA
| |
Collapse
|
28
|
Ganci M, Suleyman E, Butt H, Ball M. Associations between self-reported psychological symptom severity and gut microbiota: further support for the microgenderome. BMC Psychiatry 2022; 22:307. [PMID: 35501777 PMCID: PMC9059404 DOI: 10.1186/s12888-022-03947-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/14/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Research into the brain-gut-microbiota axis (BGMA) continues to reveal associations between gut microbiota (GM) and psychological symptom expression, inspiring new ways of conceptualising psychological disorders. However, before GM modulation can be touted as a possible auxiliary treatment option, more research is needed as inconsistencies in previous findings regarding these associations are prevalent. Additionally, the concept of the microgenderome, which proposes that GM may interact with sex hormones, has received limited attention in studies using human samples to date. However, such research has demonstrated sex specific associations between GM and psychological symptom expression. METHOD This cross-sectional retrospective study explores associations between GM species (identified through faecal microbial analysis) and symptom severity across four psychological domains (Depressive, Neurocognitive, Stress and Anxiety, and Sleep and Fatigue) for males (N = 1143) and females (N = 3467) separately. RESULTS GM species from several genera including Bifidobacterium, Clostridium, Enterococcus, and Leuconostoc were found to be differentially associated with psychological symptom severity for males and females. As such, the findings of the current study provide support for the concept of the microgenderome. CONCLUSION While further research is needed before their implementation in psychological treatment plans, the current findings suggest that modulation of GM at the species level may hold promise as auxiliary diagnostic or treatment options. These findings may give further insight into a client's presenting problem from a more holistic, multidisciplinary perspective. The clear sex divergence in associations between GM and symptoms give insight into sex discrepancies in susceptibility to psychological disorders.
Collapse
Affiliation(s)
- Michael Ganci
- Psychology Department, Institute for Health and Sport, Victoria University, PO Box 14428, Melbourne, VIC, 8001, Australia.
| | - Emra Suleyman
- grid.1019.90000 0001 0396 9544Psychology Department, Institute for Health and Sport, Victoria University, PO Box 14428, Melbourne, VIC 8001 Australia
| | - Henry Butt
- grid.1019.90000 0001 0396 9544Psychology Department, Institute for Health and Sport, Victoria University, PO Box 14428, Melbourne, VIC 8001 Australia ,Bioscreen Yarraville (Aust) Pty Ltd, Melbourne, VIC Australia
| | - Michelle Ball
- grid.1019.90000 0001 0396 9544Psychology Department, Institute for Health and Sport, Victoria University, PO Box 14428, Melbourne, VIC 8001 Australia
| |
Collapse
|
29
|
Effah F, de Gusmão Taveiros Silva NK, Vijayanathan K, Camarini R, Joly F, Taiwo B, Rabot S, Champeil-Potokar G, Bombail V, Bailey A. SEX-DEPENDENT IMPACT OF MICROBIOTA STATUS ON CEREBRAL μ -OPIOID RECEPTOR DENSITY IN FISCHER RATS. Eur J Neurosci 2022; 55:1917-1933. [PMID: 35393704 PMCID: PMC9324823 DOI: 10.1111/ejn.15666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/08/2022] [Accepted: 03/29/2022] [Indexed: 11/29/2022]
Abstract
μ‐opioid receptors (MOPr) play a critical role in social play, reward and pain, in a sex‐ and age‐dependent manner. There is evidence to suggest that sex and age differences in brain MOPr density may be responsible for this variability; however, little is known about the factors driving these differences in cerebral MOPr density. Emerging evidence highlights gut microbiota's critical influence and its bidirectional interaction with the brain on neurodevelopment. Therefore, we aimed to determine the impact of gut microbiota on MOPr density in male and female brains at different developmental stages. Quantitative [3H]DAMGO autoradiographic binding was carried out in the forebrain of male and female conventional (CON) and germ‐free (GF) rats at postnatal days (PND) 8, 22 and 116–150. Significant ‘microbiota status X sex’, ‘age X brain region’ interactions and microbiota status‐ and age‐dependent effects on MOPr binding were uncovered. Microbiota status influenced MOPr levels in males but not females, with higher MOPr levels observed in GF versus CON rats overall regions and age groups. In contrast, no overall sex differences were observed in GF or CON rats. Interestingly, within‐age planned comparison analysis conducted in frontal cortical and brain regions associated with reward revealed that this microbiota effect was restricted only to PND22 rats. Thus, this pilot study uncovers the critical sex‐dependent role of gut microbiota in regulating cerebral MOPr density, which is restricted to the sensitive developmental period of weaning. This may have implications in understanding the importance of microbiota during early development on opioid signalling and associated behaviours.
Collapse
Affiliation(s)
- Felix Effah
- Pharmacology Section, St George's University of London, Cranmer Terrace, SW17 0RE, London, UK
| | | | - Katie Vijayanathan
- Pharmacology Section, St George's University of London, Cranmer Terrace, SW17 0RE, London, UK
| | - Rosana Camarini
- Pharmacology Department, Universidade de Sao Paulo, São Paulo, Brazil
| | - Fatima Joly
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Benjamin Taiwo
- Pharmacology Section, St George's University of London, Cranmer Terrace, SW17 0RE, London, UK
| | - Sylvie Rabot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | | | - Vincent Bombail
- UMR PNCA, AgroParisTech, INRAE, Université Paris-Saclay, Paris, France
| | - Alexis Bailey
- Pharmacology Section, St George's University of London, Cranmer Terrace, SW17 0RE, London, UK
| |
Collapse
|
30
|
Krishna M, Engevik M, Queliza K, Britto S, Shah R, Ruan W, Wang H, Versalovic J, Kellermayer R. Maternal Lactobacillus reuteri supplementation shifts the intestinal microbiome in mice and provides protection from experimental colitis in female offspring. FASEB Bioadv 2022; 4:109-120. [PMID: 35141475 PMCID: PMC8814561 DOI: 10.1096/fba.2021-00078] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/01/2021] [Accepted: 10/06/2021] [Indexed: 12/26/2022] Open
Abstract
The purpose of our experiment was to explore how stochastic (inter-individual variation) gut microbiome composition may link to inflammatory bowel disease (IBD) susceptibility and guide the development of a perinatal preventative probiotic. Dextran sodium sulfate (DSS) was introduced to C57BL/BJ mice to induce acute colitis as a model of IBD. Potentially protective bacteria were identified using a discovery-validation cohort approach toward stochastic DSS susceptibility. Lactobacilli (two different cocktails of L. reuteri and L. johnsonii strains) or control media were supplemented by mouth to dams prior to delivery and during lactation (i.e., perinatal probiotic). The pups were evaluated for DSS susceptibility at young adulthood. Fecal Lactobacillus was increased in the DSS-resistant mice in both the discovery and validation cohorts. Maternal supplementation of female offspring with an L. reuteri cocktail (strains 6798-1, 6798-jm, and 6798-cm) induced progressive microbiome separation and protection against colitis by young adulthood. Maternal supplementation of L. reuteri could confer protection against DSS colitis in young adult female mice. This work is the first to exploit stochastic mammalian microbiome variation to guide microbial therapeutic identification. Our findings underscore neonatal microbiome plasticity and set the stage for the potential development of perinatally deliverable protective probiotics against human IBD.
Collapse
Affiliation(s)
- Mahesh Krishna
- Johns Hopkins School of MedicineBaltimoreMarylandUSA
- Section of Pediatric GastroenterologyBaylor College of MedicineHoustonTexasUSA
| | - Melinda Engevik
- Department of Pathology & ImmunologyBaylor College of MedicineHoustonTexasUSA
| | - Karen Queliza
- Pediatric Gastroenterology, Hepatology and NutritionMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Savini Britto
- Section of Pediatric GastroenterologyBaylor College of MedicineHoustonTexasUSA
| | - Rajesh Shah
- Department of MedicineBaylor Scott and WhiteAustinTexasUSA
| | - Wenly Ruan
- Section of Pediatric GastroenterologyBaylor College of MedicineHoustonTexasUSA
| | - Hongtao Wang
- Section of Pediatric GastroenterologyBaylor College of MedicineHoustonTexasUSA
| | - James Versalovic
- Department of Pathology & ImmunologyBaylor College of MedicineHoustonTexasUSA
| | - Richard Kellermayer
- Section of Pediatric GastroenterologyBaylor College of MedicineHoustonTexasUSA
- USDA/ARS Children's Nutrition Research CenterTexas Children's HospitalHoustonTexasUSA
| |
Collapse
|
31
|
Benedetti A, Molent C, Barcik W, Papaleo F. Social behavior in 16p11.2 and 22q11.2 copy number variations: Insights from mice and humans. GENES, BRAIN, AND BEHAVIOR 2021; 21:e12787. [PMID: 34889032 PMCID: PMC9744525 DOI: 10.1111/gbb.12787] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 11/30/2022]
Abstract
Genetic 16p11.2 and 22q11.2 deletions and duplications in humans may alter behavioral developmental trajectories increasing the risk of autism and schizophrenia spectrum disorders, and of attention-deficit/hyperactivity disorder. In this review, we will concentrate on 16p11.2 and 22q11.2 deletions' effects on social functioning, beyond diagnostic categorization. We highlight diagnostic and social sub-constructs discrepancies. Notably, we contrast evidence from human studies with social profiling performed in several mouse models mimicking 16p11.2 and 22q11.2 deletion syndromes. Given the complexity of social behavior, there is a need to assess distinct social processes. This will be important to better understand the biology underlying such genetic-dependent dysfunctions, as well as to give perspective on how therapeutic strategies can be improved. Bridges and divergent points between human and mouse studies are highlighted. Overall, we give challenges and future perspectives to sort the genetics of social heterogeneity.
Collapse
Affiliation(s)
- Arianna Benedetti
- Genetics of Cognition laboratory, Neuroscience areaIstituto Italiano di TecnologiaGenoaItaly,CNRS, GREDEGUniversité Côte d'AzurNiceFrance
| | - Cinzia Molent
- Genetics of Cognition laboratory, Neuroscience areaIstituto Italiano di TecnologiaGenoaItaly,Dipartimento di Medicina Sperimentale(Di. Mes) Università degli Studi di GenovaGenoaItaly
| | - Weronika Barcik
- Genetics of Cognition laboratory, Neuroscience areaIstituto Italiano di TecnologiaGenoaItaly
| | - Francesco Papaleo
- Genetics of Cognition laboratory, Neuroscience areaIstituto Italiano di TecnologiaGenoaItaly,Department of Neurosciences and Mental HealthFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
32
|
Saturio S, Nogacka AM, Alvarado-Jasso GM, Salazar N, de los Reyes-Gavilán CG, Gueimonde M, Arboleya S. Role of Bifidobacteria on Infant Health. Microorganisms 2021; 9:2415. [PMID: 34946017 PMCID: PMC8708449 DOI: 10.3390/microorganisms9122415] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/19/2021] [Accepted: 11/21/2021] [Indexed: 12/19/2022] Open
Abstract
Bifidobacteria are among the predominant microorganisms during infancy, being a dominant microbial group in the healthy breastfed infant and playing a crucial role in newborns and infant development. Not only the levels of the Bifidobacterium genus but also the profile and quantity of the different bifidobacterial species have been demonstrated to be of relevance to infant health. Although no definitive proof is available on the causal association, reduced levels of bifidobacteria are perhaps the most frequently observed alteration of the intestinal microbiota in infant diseases. Moreover, Bifidobacterium strains have been extensively studied by their probiotic attributes. This review compiles the available information about bifidobacterial composition and function since the beginning of life, describing different perinatal factors affecting them, and their implications on different health alterations in infancy. In addition, this review gathers exhaustive information about pre-clinical and clinical studies with Bifidobacterium strains as probiotics in neonates.
Collapse
Affiliation(s)
- Silvia Saturio
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Alicja M. Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Guadalupe M. Alvarado-Jasso
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.M.N.); (G.M.A.-J.); (N.S.); (C.G.d.l.R.-G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
33
|
Synthetic Microbiomes on the Rise-Application in Deciphering the Role of Microbes in Host Health and Disease. Nutrients 2021; 13:nu13114173. [PMID: 34836426 PMCID: PMC8621464 DOI: 10.3390/nu13114173] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
The intestinal microbiota conveys significant benefits to host physiology. Although multiple chronic disorders have been associated with alterations in the intestinal microbiota composition and function, it is still unclear whether these changes are a cause or a consequence. Hence, to translate microbiome research into clinical application, it is necessary to provide a proof of causality of host–microbiota interactions. This is hampered by the complexity of the gut microbiome and many confounding factors. The application of gnotobiotic animal models associated with synthetic communities allows us to address the cause–effect relationship between the host and intestinal microbiota by reducing the microbiome complexity on a manageable level. In recent years, diverse bacterial communities were assembled to analyze the role of microorganisms in infectious, inflammatory, and metabolic diseases. In this review, we outline their application and features. Furthermore, we discuss the differences between human-derived and model-specific communities. Lastly, we highlight the necessity of generating novel synthetic communities to unravel the microbial role associated with specific health outcomes and disease phenotypes. This understanding is essential for the development of novel non-invasive targeted therapeutic strategies to control and modulate intestinal microbiota in health and disease.
Collapse
|
34
|
Engevik MA, Herrmann B, Ruan W, Engevik AC, Engevik KA, Ihekweazu F, Shi Z, Luck B, Chang-Graham AL, Esparza M, Venable S, Horvath TD, Haidacher SJ, Hoch KM, Haag AM, Schady DA, Hyser JM, Spinler JK, Versalovic J. Bifidobacterium dentium-derived y-glutamylcysteine suppresses ER-mediated goblet cell stress and reduces TNBS-driven colonic inflammation. Gut Microbes 2021; 13:1-21. [PMID: 33985416 PMCID: PMC8128206 DOI: 10.1080/19490976.2021.1902717] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Endoplasmic reticulum (ER) stress compromises the secretion of MUC2 from goblet cells and has been linked with inflammatory bowel disease (IBD). Although Bifidobacterium can beneficially modulate mucin production, little work has been done investigating the effects of Bifidobacterium on goblet cell ER stress. We hypothesized that secreted factors from Bifidobacterium dentium downregulate ER stress genes and modulates the unfolded protein response (UPR) to promote MUC2 secretion. We identified by mass spectrometry that B. dentium secretes the antioxidant γ-glutamylcysteine, which we speculate dampens ER stress-mediated ROS and minimizes ER stress phenotypes. B. dentium cell-free supernatant and γ-glutamylcysteine were taken up by human colonic T84 cells, increased glutathione levels, and reduced ROS generated by the ER-stressors thapsigargin and tunicamycin. Moreover, B. dentium supernatant and γ-glutamylcysteine were able to suppress NF-kB activation and IL-8 secretion. We found that B. dentium supernatant, γ-glutamylcysteine, and the positive control IL-10 attenuated the induction of UPR genes GRP78, CHOP, and sXBP1. To examine ER stress in vivo, we first examined mono-association of B. dentium in germ-free mice which increased MUC2 and IL-10 levels compared to germ-free controls. However, no changes were observed in ER stress-related genes, indicating that B. dentium can promote mucus secretion without inducing ER stress. In a TNBS-mediated ER stress model, we observed increased levels of UPR genes and pro-inflammatory cytokines in TNBS treated mice, which were reduced with addition of live B. dentium or γ-glutamylcysteine. We also observed increased colonic and serum levels of IL-10 in B. dentium- and γ-glutamylcysteine-treated mice compared to vehicle control. Immunostaining revealed retention of goblet cells and mucus secretion in both B. dentium- and γ-glutamylcysteine-treated animals. Collectively, these data demonstrate positive modulation of the UPR and MUC2 production by B. dentium-secreted compounds.
Collapse
Affiliation(s)
- Melinda A. Engevik
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA,Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA,CONTACT Melinda A. Engevik Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
| | - Beatrice Herrmann
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA,Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA
| | - Wenly Ruan
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA,Section of Gastroenterology, Hepatology, and Nutrition, Texas Children’s Hospital, Houston, Texas, USA
| | - Amy C. Engevik
- Department of Surgery, Vanderbilt University Medical Center, NashvilleTN, USA
| | - Kristen A. Engevik
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Faith Ihekweazu
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA,Section of Gastroenterology, Hepatology, and Nutrition, Texas Children’s Hospital, Houston, Texas, USA
| | - Zhongcheng Shi
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA,Section of Gastroenterology, Hepatology, and Nutrition, Texas Children’s Hospital, Houston, Texas, USA
| | - Berkley Luck
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA,Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA
| | | | - Magdalena Esparza
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA,Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA
| | - Susan Venable
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA,Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA
| | - Thomas D. Horvath
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA,Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA
| | - Sigmund J. Haidacher
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA,Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA
| | - Kathleen M. Hoch
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA,Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA
| | - Anthony M. Haag
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA,Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA
| | - Deborah A. Schady
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA,Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA
| | - Joseph M. Hyser
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, USA,Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer K. Spinler
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA,Department of Pathology, Texas Children’s Hospital, Houston, Texas, USA
| | - James Versalovic
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA,Section of Gastroenterology, Hepatology, and Nutrition, Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
35
|
Luck B, Horvath TD, Engevik KA, Ruan W, Haidacher SJ, Hoch KM, Oezguen N, Spinler JK, Haag AM, Versalovic J, Engevik MA. Neurotransmitter Profiles Are Altered in the Gut and Brain of Mice Mono-Associated with Bifidobacterium dentium. Biomolecules 2021; 11:1091. [PMID: 34439760 PMCID: PMC8392031 DOI: 10.3390/biom11081091] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/29/2021] [Accepted: 07/18/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Accumulating evidence indicates that the gut microbiota can synthesize neurotransmitters as well as impact host-derived neurotransmitter levels. In the past, it has been challenging to decipher which microbes influence neurotransmitters due to the complexity of the gut microbiota. METHODS To address whether a single microbe, Bifidobacterium dentium, could regulate important neurotransmitters, we examined Bifidobacteria genomes and explored neurotransmitter pathways in secreted cell-free supernatant using LC-MS/MS. To determine if B. dentium could impact neurotransmitters in vivo, we mono-associated germ-free mice with B. dentium ATCC 27678 and examined fecal and brain neurotransmitter concentrations. RESULTS We found that B. dentium possessed the enzymatic machinery to generate γ-aminobutyric acid (GABA) from glutamate, glutamine, and succinate. Consistent with the genome analysis, we found that B. dentium secreted GABA in a fully defined microbial media and elevated fecal GABA in B. dentium mono-associated mice compared to germ-free controls. We also examined the tyrosine/dopamine pathway and found that B. dentium could synthesize tyrosine, but could not generate L-dopa, dopamine, norepinephrine, or epinephrine. In vivo, we found that B. dentium mono-associated mice had elevated levels of tyrosine in the feces and brain. CONCLUSIONS These data indicate that B. dentium can contribute to in vivo neurotransmitter regulation.
Collapse
Affiliation(s)
- Berkley Luck
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (B.L.); (T.D.H.); (S.J.H.); (K.M.H.); (N.O.); (J.K.S.); (A.M.H.); (J.V.)
- Department of Pathology, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Thomas D. Horvath
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (B.L.); (T.D.H.); (S.J.H.); (K.M.H.); (N.O.); (J.K.S.); (A.M.H.); (J.V.)
- Department of Pathology, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Kristen A. Engevik
- Department of Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Wenly Ruan
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA;
- Section of Gastroenterology, Hepatology, and Nutrition, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Sigmund J. Haidacher
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (B.L.); (T.D.H.); (S.J.H.); (K.M.H.); (N.O.); (J.K.S.); (A.M.H.); (J.V.)
- Department of Pathology, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Kathleen M. Hoch
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (B.L.); (T.D.H.); (S.J.H.); (K.M.H.); (N.O.); (J.K.S.); (A.M.H.); (J.V.)
- Department of Pathology, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Numan Oezguen
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (B.L.); (T.D.H.); (S.J.H.); (K.M.H.); (N.O.); (J.K.S.); (A.M.H.); (J.V.)
- Department of Pathology, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Jennifer K. Spinler
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (B.L.); (T.D.H.); (S.J.H.); (K.M.H.); (N.O.); (J.K.S.); (A.M.H.); (J.V.)
- Department of Pathology, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Anthony M. Haag
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (B.L.); (T.D.H.); (S.J.H.); (K.M.H.); (N.O.); (J.K.S.); (A.M.H.); (J.V.)
- Department of Pathology, Texas Children’s Hospital, Houston, TX 77030, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (B.L.); (T.D.H.); (S.J.H.); (K.M.H.); (N.O.); (J.K.S.); (A.M.H.); (J.V.)
- Department of Pathology, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Melinda A. Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; (B.L.); (T.D.H.); (S.J.H.); (K.M.H.); (N.O.); (J.K.S.); (A.M.H.); (J.V.)
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
36
|
Stanton JE, Malijauskaite S, McGourty K, Grabrucker AM. The Metallome as a Link Between the "Omes" in Autism Spectrum Disorders. Front Mol Neurosci 2021; 14:695873. [PMID: 34290588 PMCID: PMC8289253 DOI: 10.3389/fnmol.2021.695873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/14/2021] [Indexed: 12/26/2022] Open
Abstract
Metal dyshomeostasis plays a significant role in various neurological diseases such as Alzheimer's disease, Parkinson's disease, Autism Spectrum Disorders (ASD), and many more. Like studies investigating the proteome, transcriptome, epigenome, microbiome, etc., for years, metallomics studies have focused on data from their domain, i.e., trace metal composition, only. Still, few have considered the links between other "omes," which may together result in an individual's specific pathologies. In particular, ASD have been reported to have multitudes of possible causal effects. Metallomics data focusing on metal deficiencies and dyshomeostasis can be linked to functions of metalloenzymes, metal transporters, and transcription factors, thus affecting the proteome and transcriptome. Furthermore, recent studies in ASD have emphasized the gut-brain axis, with alterations in the microbiome being linked to changes in the metabolome and inflammatory processes. However, the microbiome and other "omes" are heavily influenced by the metallome. Thus, here, we will summarize the known implications of a changed metallome for other "omes" in the body in the context of "omics" studies in ASD. We will highlight possible connections and propose a model that may explain the so far independently reported pathologies in ASD.
Collapse
Affiliation(s)
- Janelle E Stanton
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland
| | - Sigita Malijauskaite
- Bernal Institute, University of Limerick, Limerick, Ireland.,Department of Chemical Sciences, University of Limerick, Limerick, Ireland
| | - Kieran McGourty
- Bernal Institute, University of Limerick, Limerick, Ireland.,Department of Chemical Sciences, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - Andreas M Grabrucker
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.,Bernal Institute, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| |
Collapse
|
37
|
Lucerne KE, Osman A, Meckel KR, Kiraly DD. Contributions of neuroimmune and gut-brain signaling to vulnerability of developing substance use disorders. Neuropharmacology 2021; 192:108598. [PMID: 33965398 PMCID: PMC8220934 DOI: 10.1016/j.neuropharm.2021.108598] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/19/2021] [Accepted: 05/03/2021] [Indexed: 02/06/2023]
Abstract
Epidemiology and clinical research indicate that only a subset of people who are exposed to drugs of abuse will go on to develop a substance use disorder. Numerous factors impact individual susceptibility to developing a substance use disorder, including intrinsic biological factors, environmental factors, and interpersonal/social factors. Given the extensive morbidity and mortality that is wrought as a consequence of substance use disorders, a substantial body of research has focused on understanding the risk factors that mediate the shift from initial drug use to pathological drug use. Understanding these risk factors provides a clear path for the development of risk mitigation strategies to help reduce the burden of substance use disorders in the population. Here we will review the rapidly growing body of literature that examines the importance of interactions between the peripheral immune system, the gut microbiome, and the central nervous system (CNS) in mediating the transition to pathological drug use. While these systems had long been viewed as distinct, there is growing evidence that there is bidirectional communication between both the immune system and the gut microbiome that drive changes in neural and behavioral plasticity relevant to substance use disorders. Further, both of these systems are highly sensitive to environmental perturbations and are implicated in numerous neuropsychiatric conditions. While the field of study examining these interactions in substance use disorders is in its relative infancy, clarifying the relationship between gut-immune-brain signaling and substance use disorders has potential to improve our understanding of individual propensity to developing addiction and yield important insight into potential treatment options.
Collapse
Affiliation(s)
- Kelsey E Lucerne
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aya Osman
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine R Meckel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Drew D Kiraly
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
38
|
Barcik W, Chiacchierini G, Bimpisidis Z, Papaleo F. Immunology and microbiology: how do they affect social cognition and emotion recognition? Curr Opin Immunol 2021; 71:46-54. [PMID: 34058687 DOI: 10.1016/j.coi.2021.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/01/2021] [Indexed: 12/25/2022]
Abstract
Social interactions profoundly influence animals' life. The quality of social interactions and many everyday life decisions are determined by a proper perception, processing and reaction to others' emotions. Notably, alterations in these social processes characterize a number of neurodevelopmental disorders, including autism spectrum disorders and schizophrenia. Increasing evidences support an implication of immune system vulnerability and inflammatory processes in disparate behavioral functions and the aforementioned neurodevelopmental disorders. In this review, we show a possible unifying view on how immune responses, within and outside the brain, and the communication between the immune system and brain responses might influence emotion recognition and related social responses. In particular, we highlight the importance of combining genetics, immunology and microbiology factors in understanding social behaviors. We underline the importance of better disentangling the whole machinery between brain-immune system interactions to better address the complexity of social processes.
Collapse
Affiliation(s)
- Weronika Barcik
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genova, Italy
| | - Giulia Chiacchierini
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genova, Italy
| | - Zisis Bimpisidis
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genova, Italy
| | - Francesco Papaleo
- Genetics of Cognition Laboratory, Neuroscience Area, Istituto Italiano di Tecnologia, Genova, Italy; Fondazione IRCCS Ca'Granda Ospedale Maggiore Policlinico, Milano, Italy.
| |
Collapse
|
39
|
Engevik MA, Danhof HA, Hall A, Engevik KA, Horvath TD, Haidacher SJ, Hoch KM, Endres BT, Bajaj M, Garey KW, Britton RA, Spinler JK, Haag AM, Versalovic J. The metabolic profile of Bifidobacterium dentium reflects its status as a human gut commensal. BMC Microbiol 2021; 21:154. [PMID: 34030655 PMCID: PMC8145834 DOI: 10.1186/s12866-021-02166-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/30/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Bifidobacteria are commensal microbes of the mammalian gastrointestinal tract. In this study, we aimed to identify the intestinal colonization mechanisms and key metabolic pathways implemented by Bifidobacterium dentium. RESULTS B. dentium displayed acid resistance, with high viability over a pH range from 4 to 7; findings that correlated to the expression of Na+/H+ antiporters within the B. dentium genome. B. dentium was found to adhere to human MUC2+ mucus and harbor mucin-binding proteins. Using microbial phenotyping microarrays and fully-defined media, we demonstrated that in the absence of glucose, B. dentium could metabolize a variety of nutrient sources. Many of these nutrient sources were plant-based, suggesting that B. dentium can consume dietary substances. In contrast to other bifidobacteria, B. dentium was largely unable to grow on compounds found in human mucus; a finding that was supported by its glycosyl hydrolase (GH) profile. Of the proteins identified in B. dentium by proteomic analysis, a large cohort of proteins were associated with diverse metabolic pathways, indicating metabolic plasticity which supports colonization of the dynamic gastrointestinal environment. CONCLUSIONS Taken together, we conclude that B. dentium is well adapted for commensalism in the gastrointestinal tract.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.
- Department of Pathology, Texas Children's Hospital, Houston, TX, USA.
- Department of Regernative Medicine & Cell Biology, Medical University of South Carolina, SC, Charleston, USA.
| | - Heather A Danhof
- Department of Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Anne Hall
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Kristen A Engevik
- Department of Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Thomas D Horvath
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Sigmund J Haidacher
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Kathleen M Hoch
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Bradley T Endres
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, TX, USA
| | - Meghna Bajaj
- Department of Chemistry and Physics, and Department of Biotechnology, Alcorn State University, Lorman, MS, 39096, USA
| | - Kevin W Garey
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, TX, USA
| | - Robert A Britton
- Department of Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer K Spinler
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - Anthony M Haag
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology, Texas Children's Hospital, Houston, TX, USA
| |
Collapse
|
40
|
Liu JCW, Gorbovskaya I, Hahn MK, Müller DJ. The Gut Microbiome in Schizophrenia and the Potential Benefits of Prebiotic and Probiotic Treatment. Nutrients 2021; 13:nu13041152. [PMID: 33807241 PMCID: PMC8065775 DOI: 10.3390/nu13041152] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome (GMB) plays an important role in developmental processes and has been implicated in the etiology of psychiatric disorders. However, the relationship between GMB and schizophrenia remains unclear. In this article, we review the existing evidence surrounding the gut microbiome in schizophrenia and the potential for antipsychotics to cause adverse metabolic events by altering the gut microbiome. We also evaluate the current evidence for the clinical use of probiotic and prebiotic treatment in schizophrenia. The current data on microbiome alteration in schizophrenia remain conflicting. Longitudinal and larger studies will help elucidate the confounding effect on the microbiome. Current studies help lay the groundwork for further investigations into the role of the GMB in the development, presentation, progression and potential treatment of schizophrenia.
Collapse
Affiliation(s)
- Jonathan C. W. Liu
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON M5T 1R8, Canada; (J.C.W.L.); (I.G.); (M.K.H.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ilona Gorbovskaya
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON M5T 1R8, Canada; (J.C.W.L.); (I.G.); (M.K.H.)
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Margaret K. Hahn
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON M5T 1R8, Canada; (J.C.W.L.); (I.G.); (M.K.H.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- Banting and Best Diabetes Centre, University of Toronto, ON M5G 2C4, Canada
| | - Daniel J. Müller
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON M5T 1R8, Canada; (J.C.W.L.); (I.G.); (M.K.H.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5T 1R8, Canada
- Correspondence:
| |
Collapse
|
41
|
Leyrolle Q, Decoeur F, Briere G, Amadieu C, Quadros ARAA, Voytyuk I, Lacabanne C, Benmamar-Badel A, Bourel J, Aubert A, Sere A, Chain F, Schwendimann L, Matrot B, Bourgeois T, Grégoire S, Leblanc JG, De Moreno De Leblanc A, Langella P, Fernandes GR, Bretillon L, Joffre C, Uricaru R, Thebault P, Gressens P, Chatel JM, Layé S, Nadjar A. Maternal dietary omega-3 deficiency worsens the deleterious effects of prenatal inflammation on the gut-brain axis in the offspring across lifetime. Neuropsychopharmacology 2021; 46:579-602. [PMID: 32781459 PMCID: PMC8026603 DOI: 10.1038/s41386-020-00793-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/16/2020] [Accepted: 07/27/2020] [Indexed: 12/18/2022]
Abstract
Maternal immune activation (MIA) and poor maternal nutritional habits are risk factors for the occurrence of neurodevelopmental disorders (NDD). Human studies show the deleterious impact of prenatal inflammation and low n-3 polyunsaturated fatty acid (PUFA) intake on neurodevelopment with long-lasting consequences on behavior. However, the mechanisms linking maternal nutritional status to MIA are still unclear, despite their relevance to the etiology of NDD. We demonstrate here that low maternal n-3 PUFA intake worsens MIA-induced early gut dysfunction, including modification of gut microbiota composition and higher local inflammatory reactivity. These deficits correlate with alterations of microglia-neuron crosstalk pathways and have long-lasting effects, both at transcriptional and behavioral levels. This work highlights the perinatal period as a critical time window, especially regarding the role of the gut-brain axis in neurodevelopment, elucidating the link between MIA, poor nutritional habits, and NDD.
Collapse
Affiliation(s)
- Q. Leyrolle
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France ,Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - F. Decoeur
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - G. Briere
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France ,grid.503269.b0000 0001 2289 8198CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400 Talence, France
| | - C. Amadieu
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. R. A. A. Quadros
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - I. Voytyuk
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - C. Lacabanne
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Benmamar-Badel
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - J. Bourel
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Aubert
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Sere
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - F. Chain
- grid.460789.40000 0004 4910 6535Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - L. Schwendimann
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - B. Matrot
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - T. Bourgeois
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France
| | - S. Grégoire
- grid.462804.c0000 0004 0387 2525Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - J. G. Leblanc
- CERELA-CONICET, San Miguel de Tucuman, 4000 Tucuman, Argentina
| | | | - P. Langella
- grid.460789.40000 0004 4910 6535Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - G. R. Fernandes
- Rene Rachou Institute – Oswaldo Cruz Foundation, Belo Horizonte, MG Brazil
| | - L. Bretillon
- grid.462804.c0000 0004 0387 2525Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - C. Joffre
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - R. Uricaru
- grid.503269.b0000 0001 2289 8198CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400 Talence, France
| | - P. Thebault
- grid.503269.b0000 0001 2289 8198CNRS, Bordeaux INP, LaBRI, UMR 5800, F-33400 Talence, France
| | - P. Gressens
- Université de Paris, NeuroDiderot, Inserm, F-75019 Paris, France ,grid.13097.3c0000 0001 2322 6764Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King’s College London, King’s Health Partners, St. Thomas’ Hospital, London, SE1 7EH UK
| | - J. M. Chatel
- grid.460789.40000 0004 4910 6535Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - S. Layé
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - A. Nadjar
- grid.488493.a0000 0004 0383 684XUniversity Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| |
Collapse
|
42
|
Darch HT, Collins MK, O'Riordan KJ, Cryan JF. Microbial memories: Sex-dependent impact of the gut microbiome on hippocampal plasticity. Eur J Neurosci 2021; 54:5235-5244. [PMID: 33458858 PMCID: PMC8451864 DOI: 10.1111/ejn.15119] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 01/06/2021] [Accepted: 01/06/2021] [Indexed: 02/06/2023]
Abstract
Germ‐free rodents, raised in the absence of a measurable gut microbiome, have been a key model to study the microbiome‐gut‐brain axis. Germ‐free mice exhibit marked behavioural and neurochemical differences to their conventionally raised counterparts. It is as yet unclear how these neurochemical differences lead to the behavioural differences. Here, we test the electrophysiological properties of hippocampal plasticity in adult germ‐free mice and compare them to conventionally raised counterparts. Whilst basal synaptic efficacy and pre‐synaptic short‐term plasticity appear normal, we find a striking alteration of hippocampal long‐term potentiation specifically in male germ‐free slices. However, the spike output of these neurons remains normal along with altered input‐output coupling, potentially indicating homeostatic compensatory mechanisms, or an altered excitation/inhibition balance. To our knowledge this is the first time the electrophysiological properties of the hippocampus have been assessed in a microbiome deficient animal. Our data indicate that the absence of a microbiome alters integration of dendritic signalling in the CA1 region in mice.
Collapse
Affiliation(s)
- Henry T Darch
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | | | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Anatomy & Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
43
|
Markey L, Pugliese A, Tian T, Roy F, Lee K, Kumamoto CA. Decreased Ecological Resistance of the Gut Microbiota in Response to Clindamycin Challenge in Mice Colonized with the Fungus Candida albicans. mSphere 2021; 6:e00982-20. [PMID: 33472981 PMCID: PMC7845615 DOI: 10.1128/msphere.00982-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023] Open
Abstract
The mammalian gut microbiota is a complex community of microorganisms which typically exhibits remarkable stability. As the gut microbiota has been shown to affect many aspects of host health, the molecular keys to developing and maintaining a "healthy" gut microbiota are highly sought after. Yet, the qualities that define a microbiota as healthy remain elusive. We used the ability to resist change in response to antibiotic disruption, a quality we refer to as ecological resistance, as a metric for the health of the bacterial microbiota. Using a mouse model, we found that colonization with the commensal fungus Candida albicans decreased the ecological resistance of the bacterial microbiota in response to the antibiotic clindamycin such that increased microbiota disruption was observed in C. albicans-colonized mice compared to that in uncolonized mice. C. albicans colonization resulted in decreased alpha diversity and small changes in abundance of bacterial genera prior to clindamycin challenge. Strikingly, co-occurrence network analysis demonstrated that C. albicans colonization resulted in sweeping changes to the co-occurrence network structure, including decreased modularity and centrality and increased density. Thus, C. albicans colonization resulted in changes to the bacterial microbiota community and reduced its ecological resistance.IMPORTANCECandida albicans is the most common fungal member of the human gut microbiota, yet its ability to interact with and affect the bacterial gut microbiota is largely uncharacterized. Previous reports showed limited changes in microbiota composition as defined by bacterial species abundance as a consequence of C. albicans colonization. We also observed only a few bacterial genera that were significantly altered in abundance in C. albicans-colonized mice; however, C. albicans colonization significantly changed the structure of the bacterial microbiota co-occurrence network. Additionally, C. albicans colonization changed the response of the bacterial microbiota ecosystem to a clinically relevant perturbation, challenge with the antibiotic clindamycin.
Collapse
Affiliation(s)
- Laura Markey
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Antonia Pugliese
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Theresa Tian
- Department of Chemical and Biological Engineering, Tufts University School of Engineering, Medford, Massachusetts, USA
| | - Farrah Roy
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kyongbum Lee
- Department of Chemical and Biological Engineering, Tufts University School of Engineering, Medford, Massachusetts, USA
| | - Carol A Kumamoto
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Ronan V, Yeasin R, Claud EC. Childhood Development and the Microbiome-The Intestinal Microbiota in Maintenance of Health and Development of Disease During Childhood Development. Gastroenterology 2021; 160:495-506. [PMID: 33307032 PMCID: PMC8714606 DOI: 10.1053/j.gastro.2020.08.065] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/25/2020] [Accepted: 08/29/2020] [Indexed: 12/11/2022]
Abstract
The composition of the intestinal microbiome affects health from the prenatal period throughout childhood, and many diseases have been associated with dysbiosis. The gut microbiome is constantly changing, from birth throughout adulthood, and several variables affect its development and content. Features of the intestinal microbiota can affect development of the brain, immune system, and lungs, as well as body growth. We review the development of the gut microbiome, proponents of dysbiosis, and interactions of the microbiota with other organs. The gut microbiome should be thought of as an organ system that has important effects on childhood development. Dysbiosis has been associated with diseases in children and adults, including autism, attention deficit hyperactivity disorder, asthma, and allergies.
Collapse
Affiliation(s)
- Victoria Ronan
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
| | - Rummanu Yeasin
- Department of Pediatrics, The University of Chicago, Chicago, Illinois; Windsor University School of Medicine, Cayon, St Kitts, West Indies
| | - Erika C Claud
- Department of Pediatrics, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
45
|
Choi TY, Choi YP, Koo JW. Mental Disorders Linked to Crosstalk between The Gut Microbiome and The Brain. Exp Neurobiol 2020; 29:403-416. [PMID: 33139585 PMCID: PMC7788310 DOI: 10.5607/en20047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/11/2022] Open
Abstract
Often called the second brain, the gut communicates extensively with the brain and vice versa. The conversation between these two organs affects a variety of physiological mechanisms that are associated with our mental health. Over the past decade, a growing body of evidence has suggested that the gut microbiome builds a unique ecosystem inside the gastrointestinal tract to maintain the homeostasis and that compositional changes in the gut microbiome are highly correlated with several mental disorders. There are ongoing efforts to treat or prevent mental disorders by regulating the gut microbiome using probiotics. These attempts are based on the seminal findings that probiotics can control the gut microbiome and affect mental conditions. However, some issues have yet to be conclusively addressed, especially the causality between the gut microbiome and mental disorders. In this review, we focus on the mechanisms by which the gut microbiome affects mental health and diseases. Furthermore, we discuss the potential use of probiotics as therapeutic agents for psychiatric disorders.
Collapse
Affiliation(s)
- Tae-Yong Choi
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| | - Young Pyo Choi
- Laboratory Animal Center, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| | - Ja Wook Koo
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| |
Collapse
|
46
|
Vogel SC, Brito NH, Callaghan BL. Early Life Stress and the Development of the Infant Gut Microbiota: Implications for Mental Health and Neurocognitive Development. Curr Psychiatry Rep 2020; 22:61. [PMID: 32918633 DOI: 10.1007/s11920-020-01186-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW We review the state of the literature examining associations between early life stress (ELS), gut microbiota, and neurocognitive development and mental health in animals and humans. We identify gaps in current models and areas for future research. RECENT FINDINGS ELS is associated with changes in gut microbiota, which correspond to changes in affective and cognitive functioning in both animals and humans. Some of these ELS-induced psychological changes can be remedied by supplementation with probiotics in early life, suggesting a potential area for intervention for ELS-exposed children. Prenatal stress exposure is rarely studied in humans in relation to gut microbiota, but animal work has suggested important associations between prenatal stress and fetal programming that should be tested in humans. The gut microbiota plays an important role in the association between ELS, neurocognitive development, and mental health. More work is needed to fully understand these associations in humans.
Collapse
Affiliation(s)
- Sarah C Vogel
- Department of Applied Psychology, New York University, 246 Greene Street, Kimball Hall, New York, NY, 10003, USA.
| | - Natalie H Brito
- Department of Applied Psychology, New York University, 246 Greene Street, Kimball Hall, New York, NY, 10003, USA
| | | |
Collapse
|
47
|
Engevik MA, Luck B, Visuthranukul C, Ihekweazu FD, Engevik AC, Shi Z, Danhof HA, Chang-Graham AL, Hall A, Endres BT, Haidacher SJ, Horvath TD, Haag AM, Devaraj S, Garey KW, Britton RA, Hyser JM, Shroyer NF, Versalovic J. Human-Derived Bifidobacterium dentium Modulates the Mammalian Serotonergic System and Gut-Brain Axis. Cell Mol Gastroenterol Hepatol 2020; 11:221-248. [PMID: 32795610 PMCID: PMC7683275 DOI: 10.1016/j.jcmgh.2020.08.002] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS The human gut microbiota can regulate production of serotonin (5-hydroxytryptamine [5-HT]) from enterochromaffin cells. However, the mechanisms underlying microbial-induced serotonin signaling are not well understood. METHODS Adult germ-free mice were treated with sterile media, live Bifidobacterium dentium, heat-killed B dentium, or live Bacteroides ovatus. Mouse and human enteroids were used to assess the effects of B dentium metabolites on 5-HT release from enterochromaffin cells. In vitro and in vivo short-chain fatty acids and 5-HT levels were assessed by mass spectrometry. Expression of tryptophan hydroxylase, short-chain fatty acid receptor free fatty acid receptor 2, 5-HT receptors, and the 5-HT re-uptake transporter (serotonin transporter) were assessed by quantitative polymerase chain reaction and immunostaining. RNA in situ hybridization assessed 5-HT-receptor expression in the brain, and 5-HT-receptor-dependent behavior was evaluated using the marble burying test. RESULTS B dentium mono-associated mice showed increased fecal acetate. This finding corresponded with increased intestinal 5-HT concentrations and increased expression of 5-HT receptors 2a, 4, and serotonin transporter. These effects were absent in B ovatus-treated mice. Application of acetate and B dentium-secreted products stimulated 5-HT release in mouse and human enteroids. In situ hybridization of brain tissue also showed significantly increased hippocampal expression of 5-HT-receptor 2a in B dentium-treated mice relative to germ-free controls. Functionally, B dentium colonization normalized species-typical repetitive and anxiety-like behaviors previously shown to be linked to 5-HT-receptor 2a. CONCLUSIONS These data suggest that B dentium, and the bacterial metabolite acetate, are capable of regulating key components of the serotonergic system in multiple host tissues, and are associated with a functional change in adult behavior.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Berkley Luck
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Chonnikant Visuthranukul
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas; Department of Pediatrics, Pediatric Nutrition Special Task Force for Activating Research (STAR), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Faith D Ihekweazu
- Pediatric Gastroenterology, Hepatology and Nutrition, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Amy C Engevik
- Department of Surgical Sciences, Vanderbilt University Medical Center, Nashville Tennessee
| | - Zhongcheng Shi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Heather A Danhof
- Department of Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | | | - Anne Hall
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas; Department of Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Bradley T Endres
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas
| | - Sigmund J Haidacher
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Thomas D Horvath
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Anthony M Haag
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Sridevi Devaraj
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Kevin W Garey
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas
| | - Robert A Britton
- Department of Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Joseph M Hyser
- Department of Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Noah F Shroyer
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas.
| |
Collapse
|
48
|
Johnson KVA, Burnet PWJ. Opposing effects of antibiotics and germ-free status on neuropeptide systems involved in social behaviour and pain regulation. BMC Neurosci 2020; 21:32. [PMID: 32698770 PMCID: PMC7374917 DOI: 10.1186/s12868-020-00583-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 07/07/2020] [Indexed: 12/16/2022] Open
Abstract
Background Recent research has revealed that the community of microorganisms inhabiting the gut affects brain development, function and behaviour. In particular, disruption of the gut microbiome during critical developmental windows can have lasting effects on host physiology. Both antibiotic exposure and germ-free conditions impact the central nervous system and can alter multiple aspects of behaviour. Social impairments are typically displayed by antibiotic-treated and germ-free animals, yet there is a lack of understanding of the underlying neurobiological changes. Since the μ-opioid, oxytocin and vasopressin systems are key modulators of mammalian social behaviour, here we investigate the effect of experimentally manipulating the gut microbiome on the expression of these pathways. Results We show that social neuropeptide signalling is disrupted in germ-free and antibiotic-treated mice, which may contribute to the behavioural deficits observed in these animal models. The most notable finding is the reduction in neuroreceptor gene expression in the frontal cortex of mice administered an antibiotic cocktail post-weaning. Additionally, the changes observed in germ-free mice were generally in the opposite direction to the antibiotic-treated mice. Conclusions Antibiotic treatment when young can impact brain signalling pathways underpinning social behaviour and pain regulation. Since antibiotic administration is common in childhood and adolescence, our findings highlight the potential adverse effects that antibiotic exposure during these key neurodevelopmental periods may have on the human brain, including the possible increased risk of neuropsychiatric conditions later in life. In addition, since antibiotics are often considered a more amenable alternative to germ-free conditions, our contrasting results for these two treatments suggest that they should be viewed as distinct models.
Collapse
Affiliation(s)
- Katerina V A Johnson
- Department of Experimental Psychology, University of Oxford, Radcliffe Observatory Quarter, Oxford, OX2 6GG, UK. .,Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX, UK.
| | - Philip W J Burnet
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, OX3 7JX, UK
| |
Collapse
|
49
|
Cheng R, Xu T, Zhang Y, Wang F, Zhao L, Jiang Y, He F. Lactobacillus rhamnosus GG and Bifidobacterium bifidum TMC3115 Can Affect Development of Hippocampal Neurons Cultured In Vitro in a Strain-Dependent Manner. Probiotics Antimicrob Proteins 2020; 12:589-599. [PMID: 31286435 DOI: 10.1007/s12602-019-09571-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
This study examined whether Lactobacillus rhamnosus GG (LGG) and Bifidobacterium bifidum TMC3115 (TMC3115) could morphologically or physiologically influence hippocampal neuronal development in vitro. Hippocampal neurons cultured in vitro were exposed to live or heat-inactivated LGG or TMC3115 for either 6 or 24 h. Neuronal morphological changes and drebrin (DRB) and synaptophysin (SYP) protein levels were monitored using immunofluorescence. And the levels of DRB, SYP, and brain-derived neurotrophic factor (BDNF), and cAMP-response element binding protein (CREB) mRNA were detected using RT-PCR. The BDNF, CREB, and phosphorylated-CREB (P-CREB) protein levels were detected by extraction-enzyme-linked immunosorbent assay (ELISA) or Western blot assays. Heat-inactivated LGG and TMC3115 could enhance neuron viability, DRB and SYP protein levels, and BDNF mRNA level were significantly altered after exposure to the tested bacteria with 6 h or 24 h. There were no significant differences in neuronal morphology or DRB, SYP, or CREB mRNA levels among the groups following bacterial exposure. However, following exposure of live TMC3115 for 24 h, the neuronal BDNF and P-CREB protein levels were both significantly up-regulated as detected by western blot assays. These results demonstrated that LGG and TMC3115 could affect neuronal viability, along with hippocampal synaptic and functional development, in a strain-dependent manner, which may also be closely associated with the physiological and culture conditions of each strain. Up-regulated P-CREB may be one of the underlying mechanisms by which the bacteria, especially neurons following exposure of live TMC3115 for 24 h, are able to regulate neuronal BDNF protein production.
Collapse
Affiliation(s)
- Ruyue Cheng
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and West China Fourth Hospital, and Healthy Food Evaluation Research Center, Sichuan University, No. 16, 3rd section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, 300050, People's Republic of China
| | - Tong Xu
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, 300050, People's Republic of China
| | - Yujie Zhang
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and West China Fourth Hospital, and Healthy Food Evaluation Research Center, Sichuan University, No. 16, 3rd section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, 300050, People's Republic of China
| | - Feng Wang
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, 300050, People's Republic of China
| | - Linsen Zhao
- Hebei Inatural Biotech Co. Ltd., Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Yugang Jiang
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, 300050, People's Republic of China.
| | - Fang He
- Department of Nutrition, Food Hygiene and Toxicology, West China School of Public Health and West China Fourth Hospital, and Healthy Food Evaluation Research Center, Sichuan University, No. 16, 3rd section, South Renmin Road, Wuhou District, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
50
|
Luck B, Engevik MA, Ganesh BP, Lackey EP, Lin T, Balderas M, Major A, Runge J, Luna RA, Sillitoe RV, Versalovic J. Bifidobacteria shape host neural circuits during postnatal development by promoting synapse formation and microglial function. Sci Rep 2020; 10:7737. [PMID: 32385412 PMCID: PMC7210968 DOI: 10.1038/s41598-020-64173-3] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 04/12/2020] [Indexed: 12/17/2022] Open
Abstract
We hypothesized that early-life gut microbiota support the functional organization of neural circuitry in the brain via regulation of synaptic gene expression and modulation of microglial functionality. Germ-free mice were colonized as neonates with either a simplified human infant microbiota consortium consisting of four Bifidobacterium species, or with a complex, conventional murine microbiota. We examined the cerebellum, cortex, and hippocampus of both groups of colonized mice in addition to germ-free control mice. At postnatal day 4 (P4), conventionalized mice and Bifidobacterium-colonized mice exhibited decreased expression of synapse-promoting genes and increased markers indicative of reactive microglia in the cerebellum, cortex and hippocampus relative to germ-free mice. By P20, both conventional and Bifidobacterium-treated mice exhibited normal synaptic density and neuronal activity as measured by density of VGLUT2+ puncta and Purkinje cell firing rate respectively, in contrast to the increased synaptic density and decreased firing rate observed in germ-free mice. The conclusions from this study further reveal how bifidobacteria participate in establishing functional neural circuits. Collectively, these data indicate that neonatal microbial colonization of the gut elicits concomitant effects on the host CNS, which promote the homeostatic developmental balance of neural connections during the postnatal time period.
Collapse
Affiliation(s)
- Berkley Luck
- Department of Pathology, Texas Children's Hospital, Houston, Texas, United States of America
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Integrative Molecular and Biomedical Sciences (IMBS), Baylor College of Medicine, Houston, Texas, United States of America
| | - Melinda A Engevik
- Department of Pathology, Texas Children's Hospital, Houston, Texas, United States of America.
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America.
| | - Bhanu Priya Ganesh
- Department of Neurology, University of Texas Health Science Center, Houston, Texas, United States of America
| | - Elizabeth P Lackey
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
| | - Tao Lin
- Department of Pathology, Texas Children's Hospital, Houston, Texas, United States of America
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Miriam Balderas
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Microbiome Center, Texas Children's Hospital, Houston, Texas, United States of America
| | - Angela Major
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jessica Runge
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ruth Ann Luna
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Roy V Sillitoe
- Department of Pathology, Texas Children's Hospital, Houston, Texas, United States of America
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, United States of America
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - James Versalovic
- Department of Pathology, Texas Children's Hospital, Houston, Texas, United States of America
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, United States of America
- Texas Children's Microbiome Center, Texas Children's Hospital, Houston, Texas, United States of America
| |
Collapse
|