1
|
Shi YC, Wu SC, Lin YC, Zheng YJ, Huang CH, Lee BH. Development of fermented Atemoya (Annona cherimola × Annona squamosa)-Amazake increased intestinal next-generation probiotics. Food Chem 2024; 459:140373. [PMID: 38986198 DOI: 10.1016/j.foodchem.2024.140373] [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: 04/17/2024] [Revised: 06/26/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Akkermansia muciniphila and Faecalibacterium prausnitzii are next-generation probiotics, which has been reported to protect disease and effectively utilize various carbohydrates (starch and pectin) as nutrients for growth. Atemoya exhibiting fruity flavor, which is suitable for enhancing aroma and attenuating unpleasant taste caused by the koji metabolites. Results indicated that malic acid was increased (from 42.4 to 70.1 mg/100 g) in fermented Atemoya-Amazake. In addition, fermented Atemoya-Amazake elevated growthes in A. muciniphila and F. prausnitzii. Similarly, the populations of Parabacteroides (5.7 fold) and Akkermansia (1.66 fold) were elevated by fermented Atemoya-Amazake treatment in an in vitro simulated gastrointestinal system compared to the control group. Results revealed that fermented Atemoya-Amazake modulated the intestinal microbiota through increasing the production of short-chain fatty acids (exhibiting anti-pathogenic activity) for 2.1, 2.5, 2.6, and 2.1 folds in acetic acid, propionic acid, isobutyric acid, and butyric acid, respectively; suggesting this fermented Atemoya-Amazake could be applied in intestinal protection.
Collapse
Affiliation(s)
- Yeu-Ching Shi
- Department of Food Sciences, National Chiayi University, Chiayi, Taiwan
| | - She-Ching Wu
- Department of Food Sciences, National Chiayi University, Chiayi, Taiwan.
| | - Yi-Ching Lin
- Department of Horticultural Science, National Chiayi University, Chiayi, Taiwan
| | - Yu-Juan Zheng
- Department of Horticultural Science, National Chiayi University, Chiayi, Taiwan
| | - Cheng-Hao Huang
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Bao-Hong Lee
- Department of Horticultural Science, National Chiayi University, Chiayi, Taiwan.
| |
Collapse
|
2
|
Meadows V, Antonio JM, Ferraris RP, Gao N. Ruminococcus gnavus in the gut: driver, contributor, or innocent bystander in steatotic liver disease? FEBS J 2024:e17327. [PMID: 39589934 DOI: 10.1111/febs.17327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/29/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024]
Abstract
The human gut microbiome plays a crucial role in regulating intestinal and systemic health, impacting host immune response and metabolic function. Dysbiosis of the gut microbiome is linked to various diseases, including steatotic liver diseases. Metabolic dysfunction-associated steatotic liver disease (MASLD), a chronic liver disease characterized by excess hepatic lipid content and impaired metabolism, is the leading cause of liver disease worldwide. Among the gut microbes, Ruminococcus gnavus (R. gnavus) has garnered attention for its association with inflammatory and metabolic diseases. While R. gnavus abundance correlates to liver fat accumulation, further research is needed to identify a causal role or therapeutic intervention in steatotic liver disease. This review surveys our current understanding of R. gnavus in the development and progression of steatotic liver diseases, highlighting its potential mechanisms through metabolite secretion, and emphasizes the need for comprehensive microbiome analyses and longitudinal studies to better understand R. gnavus' impact on liver health. This knowledge could pave the way for targeted interventions aimed at modulating gut microbiota to treat and prevent MASLD and its comorbidities.
Collapse
Affiliation(s)
- Vik Meadows
- Department of Biological Sciences, School of Arts & Sciences, Rutgers University, Newark, NJ, USA
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Jayson M Antonio
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Ronaldo P Ferraris
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Nan Gao
- Department of Biological Sciences, School of Arts & Sciences, Rutgers University, Newark, NJ, USA
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| |
Collapse
|
3
|
Ma X, Li M, Wang X, Qi G, Wei L, Zhang D. Sialylation in the gut: From mucosal protection to disease pathogenesis. Carbohydr Polym 2024; 343:122471. [PMID: 39174097 DOI: 10.1016/j.carbpol.2024.122471] [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: 05/07/2024] [Revised: 06/19/2024] [Accepted: 07/07/2024] [Indexed: 08/24/2024]
Abstract
Sialylation, a crucial post-translational modification of glycoconjugates, entails the attachment of sialic acid (SA) to the terminal glycans of glycoproteins and glycolipids through a tightly regulated enzymatic process involving various enzymes. This review offers a comprehensive exploration of sialylation within the gut, encompassing its involvement in mucosal protection and its impact on disease progression. The sialylation of mucins and epithelial glycoproteins contributes to the integrity of the intestinal mucosal barrier. Furthermore, sialylation regulates immune responses in the gut, shaping interactions among immune cells, as well as their activation and tolerance. Additionally, the gut microbiota and gut-brain axis communication are involved in the role of sialylation in intestinal health. Altered sialylation patterns have been implicated in various intestinal diseases, including inflammatory bowel disease (IBD), colorectal cancer (CRC), and other intestinal disorders. Emerging research underscores sialylation as a promising avenue for diagnostic, prognostic, and therapeutic interventions in intestinal diseases. Potential strategies such as sialic acid supplementation, inhibition of sialidases, immunotherapy targeting sialylated antigens, and modulation of sialyltransferases have been utilized in the treatment of intestinal diseases. Future research directions will focus on elucidating the molecular mechanisms underlying sialylation alterations, identifying sialylation-based biomarkers, and developing targeted interventions for precision medicine approaches.
Collapse
Affiliation(s)
- Xueni Ma
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Muyang Li
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xiaochun Wang
- Department of Gastroenterology, Gansu Provincial Hospital, Lanzhou, China
| | - Guoqing Qi
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lina Wei
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China; Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
4
|
Xiang Q, Yu M, Cai Q, Hu M, Rao B, Liang X, Liu Z, Xie Y, Cen K, Zhang R, Xu H, Liu Y. Multi-omics insights into the microbiota-gut-brain axis and cognitive improvement post-bariatric surgery. J Transl Med 2024; 22:945. [PMID: 39420319 PMCID: PMC11484437 DOI: 10.1186/s12967-024-05757-9] [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: 06/03/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Although numerous studies have shown that bariatric surgery results in sustained weight loss and modifications in gut microbiota composition and cognitive function, the exact underlying mechanisms are unclear. This study aimed to investigate the effects of bariatric surgery on cognitive function through the microbiota-gut-brain axis (MGBA). METHODS Demographic data, serum samples, fecal samples, cognitive assessment scales, and resting-state functional connectivity magnetic resonance imaging (rs-fMRI) scans were obtained from 39 obese patients before and after (6 months) laparoscopic sleeve gastrectomy (LSG). PCA analysis, OPLS-DA analysis, and permutation tests were used to conduct fecal 16 S microbiota profiling, serum metabolomics, and neuroimaging analyses, and a bariatric surgery-specific rs-fMRI brain functional connectivity network was constructed. Spearman correlation analysis and Co-inertia analysis were employed to correlate significant alterations in cognitive assessment scales and resting-state functional connectivity difference networks with differential serum metabolites and 16 S microbiota data to identify key gut microbiota and serum metabolic factors. RESULTS LSG significantly reduced the weight of obese patients, with reductions of up to 28%. Furthermore, cognitive assessment scale measurements revealed that LSG enhanced cognitive functions, including memory (HVLT, p = 0.000) and executive function (SCWT, p = 0.008). Also, LSG significantly altered gut microbiota composition (p = 0.001), with increased microbial abundance and diversity (p < 0.05). Moreover, serum metabolite levels were significantly altered, revealing intergroup differences in 229 metabolites mapped to 72 metabolic pathways (p < 0.05, VIP > 1). Spearman correlation analysis among cognitive assessment scales, gut microbiota species, and serum metabolites revealed correlations with 68 gut microbiota species and 138 serum metabolites (p < 0.05). Furthermore, pairwise correlations were detected between gut microbiota and serum metabolites (p < 0.05). Functional neuroimaging analysis revealed that LSG increased functional connectivity in cognitive-related frontotemporal networks (FPN, p < 0.01). Additionally, normalization of the default mode network (DMN) and salience network (SN) connectivity was observed after LSG (p < 0.001). Further canonical correlation and correlation analysis suggested that the cognitive-related brain network changes induced by LSG were associated with key gut microbiota species (Akkermansia, Blautia, Collinsella, Phascolarctobacterium, and Ruminococcus, p < 0.05) and neuroactive metabolites (Glycine, L-Serine, DL-Dopa, SM (d18:1/24:1(15Z), p < 0.05). CONCLUSION These findings indicate the pathophysiological role of the microbiota-gut-brain axis in enhancing cognitive function after bariatric surgery, and the study provides a basis for clinical dietary adjustments, probiotic supplementation, and guidance for bariatric surgery, but further research is still needed. TRIAL REGISTRATION Chinese Clinical Trial Registry, ChiCTR2100049403. Registered 02 August 2021, https://www.chictr.org.cn/ .
Collapse
Affiliation(s)
- Qiaoyuan Xiang
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China
| | - Minhua Yu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qi Cai
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China
| | - Mengjie Hu
- Department of Hepatobiliary, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Bo Rao
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xin Liang
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China
| | - Zhenxing Liu
- Department of Neurology, Yiling Hospital of Yichang City, Yichang, Hubei, China
| | - Yu Xie
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China
| | - Kuan Cen
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China
| | - Renwei Zhang
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Yumin Liu
- Dapartment of Neurology, Zhongnan Hospital of Wuhan University, No.169 Donghu Street, Wuhan, Hubei, 430000, China.
| |
Collapse
|
5
|
Huovinen V, Aatsinki AK, Kataja EL, Munukka E, Keskitalo A, Lamichhane S, Raunioniemi P, Bridgett DJ, Lahti L, O'Mahony SM, Dickens A, Korja R, Karlsson H, Nolvi S, Karlsson L. Infant gut microbiota and negative and fear reactivity. Dev Psychopathol 2024; 36:2016-2031. [PMID: 37974473 DOI: 10.1017/s0954579423001396] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
BACKGROUND Studies indicate that gut microbiota is related to neurodevelopmental and behavioral outcomes. Accordingly, early gut microbiota composition (GMC) has been linked to child temperament, but research is still scarce. The aim of this study was to examine how early GMC at 2.5 months is associated with child negative and fear reactivity at 8 and 12 months since they are potentially important intermediate phenotypes of later child psychiatric disorders. METHODS Our study population was 330 infants enrolled in the longitudinal FinnBrain Birth Cohort Study. Gut microbiota composition was analyzed using stool sample 16s rRNA sequencing. Negative and fear reactivity were assessed using the Laboratory Temperament Assessment Battery (Lab-TAB) at child's age of 8 months (n =150) and the Infant Behavior Questionnaire-Revised Short Form (IBQ-R SF) at child's age of 12 months (n = 276). CONCLUSIONS We found a positive association between alpha diversity and reported fear reactivity and differing microbial community composition based on negative reactivity for boys. Isobutyric acid correlated with observed negative reactivity, however, this association attenuated in the linear model. Several genera were associated with the selected infant temperament traits. This study adds to the growing literature on links between infant gut microbiota and temperament informing future mechanistic studies.
Collapse
Affiliation(s)
- Venla Huovinen
- Department of Clinical Medicine, Psychiatry, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
| | - Anna-Katariina Aatsinki
- Department of Clinical Medicine, Psychiatry, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
- Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland
| | - Eeva-Leena Kataja
- Department of Clinical Medicine, Psychiatry, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
| | - Eveliina Munukka
- Microbiome Biobank, Research Center for Infections and Immunity Institute of Biomedicine, University of Turku and Turku University, Hospital, Turku, Finland
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Anniina Keskitalo
- Department of Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Santosh Lamichhane
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Peppi Raunioniemi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - David J Bridgett
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Leo Lahti
- Department of Computing, Faculty of Technology, University of Turku, Turku, Finland
| | - Siobhain M O'Mahony
- Department of Anatomy and Neuroscience, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Alex Dickens
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Riikka Korja
- Department of Clinical Medicine, Psychiatry, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
- Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
| | - Hasse Karlsson
- Department of Clinical Medicine, Psychiatry, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
- Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland
| | - Saara Nolvi
- Department of Clinical Medicine, Psychiatry, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
- Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
- Turku Institute for Advanced Studies, University of Turku, Turku, Finland
| | - Linnea Karlsson
- Department of Clinical Medicine, Psychiatry, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
- Centre for Population Health Research, Turku University Hospital and University of Turku, Turku, Finland
- Department of Clinical Medicine, Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
6
|
Flores JA, Antonio JM, Suntornsaratoon P, Meadows V, Bandyopadhyay S, Han J, Singh R, Balasubramanian I, Upadhyay R, Liu Y, Bonder EM, Kiela P, Su X, Ferraris R, Gao N. The arginine and nitric oxide metabolic pathway regulate the gut colonization and expansion of Ruminococcous gnavus. J Biol Chem 2024; 300:107614. [PMID: 39089585 PMCID: PMC11387683 DOI: 10.1016/j.jbc.2024.107614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 08/04/2024] Open
Abstract
Ruminococcus gnavus is a mucolytic commensal bacterium whose increased gut colonization has been associated with chronic inflammatory and metabolic diseases in humans. Whether R. gnavus metabolites can modulate host intestinal physiology remains largely understudied. We performed untargeted metabolomic and bulk RNA-seq analyses using R. gnavus monocolonization in germ-free mice. Based on transcriptome-metabolome correlations, we tested the impact of specific arginine metabolites on intestinal epithelial production of nitric oxide (NO) and examined the effect of NO on the growth of various strains of R. gnavus in vitro and in nitric oxide synthase 2 (Nos2)-deficient mice. R. gnavus produces specific arginine, tryptophan, and tyrosine metabolites, some of which are regulated by the environmental richness of sialic acid and mucin. R. gnavus colonization promotes expression of amino acid transporters and enzymes involved in metabolic flux of arginine and associated metabolites into NO. R. gnavus induced elevated levels of NOS2, while Nos2 ablation resulted in R. gnavus expansion in vivo. The growth of various R. gnavus strains can be inhibited by NO. Specific R. gnavus metabolites modulate intestinal epithelial cell NOS2 abundance and reduce epithelial barrier function at higher concentrations. Intestinal colonization and interaction with R. gnavus are partially regulated by an arginine-NO metabolic pathway, whereby a balanced control by the gut epithelium may restrain R. gnavus growth in healthy individuals. Disruption in this arginine metabolic regulation will contribute to the expansion and blooming of R. gnavus.
Collapse
Affiliation(s)
- Juan A Flores
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Jayson M Antonio
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Panan Suntornsaratoon
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Vik Meadows
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA; Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | | | - Jiangmeng Han
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA; Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Rajbir Singh
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | | | - Ravij Upadhyay
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Yue Liu
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Pawel Kiela
- Daniel Cracchiolo Institute for Pediatric Autoimmune Disease Research, Steele Children's Research Center, Department of Pediatrics, University of Arizona, Tucson, Arizona, USA
| | - Xiaoyang Su
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Ronaldo Ferraris
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA.
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA; Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA.
| |
Collapse
|
7
|
Reeves KD, Figuereo YF, Weis VG, Hsu FC, Engevik MA, Krigsman A, Walker SJ. Mapping the geographical distribution of the mucosa-associated gut microbiome in GI-symptomatic children with autism spectrum disorder. Am J Physiol Gastrointest Liver Physiol 2024; 327:G217-G234. [PMID: 38887795 PMCID: PMC11637567 DOI: 10.1152/ajpgi.00101.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/16/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by cognitive, behavioral, and communication impairments. In the past few years, it has been proposed that alterations in the gut microbiota may contribute to an aberrant communication between the gut and brain in children with ASD. Consistent with this notion, several studies have demonstrated that children with ASD have an altered fecal microbiota compared with typically developing (TD) children. However, it is unclear where along the length of the gastrointestinal (GI) tract these alterations in microbial communities occur. In addition, the variation between specific mucosa-associated communities remains unknown. To address this gap in knowledge of the microbiome associated with ASD, biopsies from the antrum, duodenum, ileum, right colon, and rectum of children with ASD and age- and sex-matched TD children were examined by 16S rRNA sequencing. We observed an overall elevated abundance of Bacillota and Bacteroidota and a decreased abundance of Pseudomonadota in all GI tract regions of both male and female children with ASD compared with TD children. Further analysis at the genera level revealed unique differences in the microbiome in the different regions of the GI tract in children with ASD compared with TD children. We also observed sex-specific differences in the gut microbiota composition in children with ASD. These data indicate that the microbiota of children with ASD is altered in multiple regions of the GI tract and that different anatomic locations have unique alterations in mucosa-associated bacterial genera.NEW & NOTEWORTHY Analysis in stool samples has shown gut microbiota alterations in children with autism spectrum disorder (ASD) compared with typically developing (TD) children. However, it is unclear which segment(s) of the gut exhibit alterations in microbiome composition. In this study, we examined microbiota composition along the gastrointestinal (GI) tract in the stomach, duodenum, ileum, right colon, and rectum. We found site-specific and sex-specific differences in the gut microbiota of children with ASD, compared with controls.
Collapse
Affiliation(s)
- Kimberly D Reeves
- Center for Precision Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina, United States
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem North Carolina, United States
| | - Yosauri F Figuereo
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Victoria G Weis
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Melinda A Engevik
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, United States
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Arthur Krigsman
- Pediatric Gastroenterology Resources, Georgetown, Texas, United States
| | - Stephen J Walker
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| |
Collapse
|
8
|
Sui Y, Feng X, Ma Y, Zou Y, Liu Y, Huang J, Zhu X, Wang J. BHBA attenuates endoplasmic reticulum stress-dependent neuroinflammation via the gut-brain axis in a mouse model of heat stress. CNS Neurosci Ther 2024; 30:e14840. [PMID: 38973202 PMCID: PMC11228358 DOI: 10.1111/cns.14840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Heat stress (HS) commonly occurs as a severe pathological response when the body's sensible temperature exceeds its thermoregulatory capacity, leading to the development of chronic brain inflammation, known as neuroinflammation. Emerging evidence suggests that HS leads to the disruption of the gut microbiota, whereas abnormalities in the gut microbiota have been demonstrated to affect neuroinflammation. However, the mechanisms underlying the effects of HS on neuroinflammation are poorly studied. Meanwhile, effective interventions have been unclear. β-Hydroxybutyric acid (BHBA) has been found to have neuroprotective and anti-inflammatory properties in previous studies. This study aims to explore the modulatory effects of BHBA on neuroinflammation induced by HS and elucidate the underlying molecular mechanisms. METHODS An in vivo and in vitro model of HS was constructed under the precondition of BHBA pretreatment. The modulatory effects of BHBA on HS-induced neuroinflammation were explored and the underlying molecular mechanisms were elucidated by flow cytometry, WB, qPCR, immunofluorescence staining, DCFH-DA fluorescent probe assay, and 16S rRNA gene sequencing of colonic contents. RESULTS Heat stress was found to cause gut microbiota disruption in HS mouse models, and TM7 and [Previotella] spp. may be the best potential biomarkers for assessing the occurrence of HS. Fecal microbiota transplantation associated with BHBA effectively reversed the disruption of gut microbiota in HS mice. Moreover, BHBA may inhibit microglia hyperactivation, suppress neuroinflammation (TNF-α, IL-1β, and IL-6), and reduce the expression of cortical endoplasmic reticulum stress (ERS) markers (GRP78 and CHOP) mainly through its modulatory effects on the gut microbiota (TM7, Lactobacillus spp., Ruminalococcus spp., and Prevotella spp.). In vitro experiments revealed that BHBA (1 mM) raised the expression of the ERS marker GRP78, enhanced cellular activity, and increased the generation of reactive oxygen species (ROS) and anti-inflammatory cytokines (IL-10), while also inhibiting HS-induced apoptosis, ROS production, and excessive release of inflammatory cytokines (TNF-α and IL-1β) in mouse BV2 cells. CONCLUSION β-Hydroxybutyric acid may be an effective agent for preventing neuroinflammation in HS mice, possibly due to its ability to inhibit ERS and subsequent microglia neuroinflammation via the gut-brain axis. These findings lay the groundwork for future research and development of BHBA as a preventive drug for HS and provide fresh insights into techniques for treating neurological illnesses by modifying the gut microbiota.
Collapse
Affiliation(s)
- Yuzhen Sui
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiao Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yue Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yimeng Zou
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yanli Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jian Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jianguo Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
9
|
Harvey DJ. Analysis of carbohydrates and glycoconjugates by matrix-assisted laser desorption/ionization mass spectrometry: An update for 2021-2022. MASS SPECTROMETRY REVIEWS 2024. [PMID: 38925550 DOI: 10.1002/mas.21873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 06/28/2024]
Abstract
The use of matrix-assisted laser desorption/ionization (MALDI) mass spectrometry for the analysis of carbohydrates and glycoconjugates is a well-established technique and this review is the 12th update of the original article published in 1999 and brings coverage of the literature to the end of 2022. As with previous review, this review also includes a few papers that describe methods appropriate to analysis by MALDI, such as sample preparation, even though the ionization method is not MALDI. The review follows the same format as previous reviews. It is divided into three sections: (1) general aspects such as theory of the MALDI process, matrices, derivatization, MALDI imaging, fragmentation, quantification and the use of computer software for structural identification. (2) Applications to various structural types such as oligo- and polysaccharides, glycoproteins, glycolipids, glycosides and biopharmaceuticals, and (3) other general areas such as medicine, industrial processes, natural products and glycan synthesis where MALDI is extensively used. Much of the material relating to applications is presented in tabular form. MALDI is still an ideal technique for carbohydrate analysis, particularly in its ability to produce single ions from each analyte and advancements in the technique and range of applications show little sign of diminishing.
Collapse
|
10
|
Víquez-R L, Henrich M, Riegel V, Bader M, Wilhelm K, Heurich M, Sommer S. A taste of wilderness: supplementary feeding of red deer (Cervus elaphus) increases individual bacterial microbiota diversity but lowers abundance of important gut symbionts. Anim Microbiome 2024; 6:28. [PMID: 38745212 PMCID: PMC11094858 DOI: 10.1186/s42523-024-00315-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 05/09/2024] [Indexed: 05/16/2024] Open
Abstract
The gut microbiome plays a crucial role in the health and well-being of animals. It is especially critical for ruminants that depend on this bacterial community for digesting their food. In this study, we investigated the effects of management conditions and supplemental feeding on the gut bacterial microbiota of red deer (Cervus elaphus) in the Bavarian Forest National Park, Germany. Fecal samples were collected from free-ranging deer, deer within winter enclosures, and deer in permanent enclosures. The samples were analyzed by high-throughput sequencing of the 16 S rRNA gene. The results showed that the gut bacterial microbiota differed in diversity, abundance, and heterogeneity within and between the various management groups. Free-ranging deer exhibited lower alpha diversity compared with deer in enclosures, probably because of the food supplementation available to the animals within the enclosures. Free-living individuals also showed the highest beta diversity, indicating greater variability in foraging grounds and plant species selection. Moreover, free-ranging deer had the lowest abundance of potentially pathogenic bacterial taxa, suggesting a healthier gut microbiome. Winter-gated deer, which spent some time in enclosures, exhibited intermediate characteristics between free-ranging and all-year-gated deer. These findings suggest that the winter enclosure management strategy, including supplementary feeding with processed plants and crops, has a significant impact on the gut microbiome composition of red deer. Overall, this study provides important insights into the effects of management conditions, particularly winter enclosure practices, on the gut microbiome of red deer. Understanding these effects is crucial for assessing the potential health implications of management strategies and highlights the value of microbiota investigations as health marker.
Collapse
Affiliation(s)
- Luis Víquez-R
- Institute of Evolutionary Ecology and Conservation Genomics, Ulm University, Ulm, Baden-Württemberg, Germany.
- Department of Biology, Bucknell University, Lewisburg, PA, USA.
| | - Maik Henrich
- Department of National Park Monitoring and Animal Management, Bavarian Forest National Park, Grafenau, Bayern, Germany
- Chair of Wildlife Ecology and Wildlife Management, University of Freiburg, Freiburg, Baden-Württemberg, Germany
| | - Vanessa Riegel
- Institute of Evolutionary Ecology and Conservation Genomics, Ulm University, Ulm, Baden-Württemberg, Germany
| | - Marvin Bader
- Institute of Evolutionary Ecology and Conservation Genomics, Ulm University, Ulm, Baden-Württemberg, Germany
- Albert-Ludwigs University, Freiburg, Baden-Württemberg, Germany
| | - Kerstin Wilhelm
- Institute of Evolutionary Ecology and Conservation Genomics, Ulm University, Ulm, Baden-Württemberg, Germany
| | - Marco Heurich
- Department of National Park Monitoring and Animal Management, Bavarian Forest National Park, Grafenau, Bayern, Germany
- Chair of Wildlife Ecology and Wildlife Management, University of Freiburg, Freiburg, Baden-Württemberg, Germany
- Institute for Forest and Wildlife Management, Inland Norway University of Applied Sciences, Koppang, NO-34, Norway
| | - Simone Sommer
- Institute of Evolutionary Ecology and Conservation Genomics, Ulm University, Ulm, Baden-Württemberg, Germany.
| |
Collapse
|
11
|
He Y, Han C, Li C, Yin X, Wang J, Gu L, Yan R, Liu B, Zhou X, He W. Role of N-acetylkynurenine in mediating the effect of gut microbiota on urinary tract infection: a Mendelian randomization study. Front Microbiol 2024; 15:1384095. [PMID: 38711967 PMCID: PMC11070472 DOI: 10.3389/fmicb.2024.1384095] [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: 02/12/2024] [Accepted: 04/02/2024] [Indexed: 05/08/2024] Open
Abstract
Introduction This study explored the causal connections between gut microbiota (GM), urinary tract infection (UTI), and potential metabolite mediators using Mendelian randomization (MR). Methods We utilized summary statistics from the most comprehensive and extensive genome-wide association studies (GWAS) available to date, including 196 bacterial traits for GM, 1,091 blood metabolites, 309 metabolite ratios, alongside UTI data from ukb-b-8814 and ebi-a-GCST90013890. Bidirectional MR analyses were conducted to investigate the causal links between GM and UTI. Subsequently, two MR analyses were performed to identify the potential mediating metabolites, followed by a two-step MR analysis to quantify the mediation proportion. Results Our findings revealed that out of the total 15 bacterial traits, significant associations with UTI risk were observed across both datasets. Particularly, taxon g_Ruminococcaceae UCG010 displayed a causal link with a diminished UTI risk in both datasets (ukb-b-8814: odds ratio [OR] = 0.9964, 95% confidence interval [CI] = 0.9930-0.9997, P = 0.036; GCST90013890: OR = 0.8252, 95% CI = 0.7217-0.9436, P = 0.005). However, no substantial changes in g_Ruminococcaceae UCG010 due to UTI were noted (ukb-b-8814: β = 0.51, P = 0.87; ebi-a-GCST90013890: β = -0.02, P = 0.77). Additionally, variations in 56 specific metabolites were induced by g_Ruminococcaceae UCG010, with N-acetylkynurenine (NAK) exhibiting a causal correlation with UTI. A negative association was found between g_Ruminococcaceae UCG010 and NAK (OR: 0.8128, 95% CI: 0.6647-0.9941, P = 0.044), while NAK was positively associated with UTI risk (OR: 1.0009; 95% CI: 1.0002-1.0016; P = 0.0173). Mediation analysis revealed that the association between g_Ruminococcaceae UCG010 and UTI was mediated by NAK with a mediation proportion of 5.07%. Discussion This MR study provides compelling evidence supporting the existence of causal relationships between specific GM taxa and UTI, along with potential mediating metabolites.
Collapse
Affiliation(s)
- Yining He
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Chao Han
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Yancheng Dafeng Hospital of Chinese Medicine, Teaching Hospital of Nanjing University of Chinese Medicine, Yancheng, China
| | - Chengjuan Li
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Xiaofan Yin
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Jiawen Wang
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Lina Gu
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Ruxue Yan
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Buhui Liu
- Department of Human Anatomy, Xuzhou Medical University, Xuzhou, China
| | - Xuan Zhou
- Department of Respiratory, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weiming He
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| |
Collapse
|
12
|
Lai Y, Tang W, Luo X, Zheng H, Zhang Y, Wang M, Yu G, Yang M. Gut microbiome and metabolome to discover pathogenic bacteria and probiotics in ankylosing spondylitis. Front Immunol 2024; 15:1369116. [PMID: 38711505 PMCID: PMC11070502 DOI: 10.3389/fimmu.2024.1369116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/01/2024] [Indexed: 05/08/2024] Open
Abstract
Objective Previous research has partially revealed distinct gut microbiota in ankylosing spondylitis (AS). In this study, we performed non-targeted fecal metabolomics in AS in order to discover the microbiome-metabolome interface in AS. Based on prospective cohort studies, we further explored the impact of the tumor necrosis factor inhibitor (TNFi) on the gut microbiota and metabolites in AS. Methods To further understand the gut microbiota and metabolites in AS, along with the influence of TNFi, we initiated a prospective cohort study. Fecal samples were collected from 29 patients with AS before and after TNFi therapy and 31 healthy controls. Metagenomic and metabolomic experiments were performed on the fecal samples; moreover, validation experiments were conducted based on the association between the microbiota and metabolites. Results A total of 7,703 species were annotated using the metagenomic sequencing system and by profiling the microbial community taxonomic composition, while 50,046 metabolites were identified using metabolite profiling. Differential microbials and metabolites were discovered between patients with AS and healthy controls. Moreover, TNFi was confirmed to partially restore the gut microbiota and the metabolites. Multi-omics analysis of the microbiota and metabolites was performed to determine the associations between the differential microbes and metabolites, identifying compounds such as oxypurinol and biotin, which were correlated with the inhibition of the pathogenic bacteria Ruminococcus gnavus and the promotion of the probiotic bacteria Bacteroides uniformis. Through experimental studies, the relationship between microbes and metabolites was further confirmed, and the impact of these two types of microbes on the enterocytes and the inflammatory cytokine interleukin-18 (IL-18) was explored. Conclusion In summary, multi-omics exploration elucidated the impact of TNFi on the gut microbiota and metabolites and proposed a novel therapeutic perspective: supplementation of compounds to inhibit potential pathogenic bacteria and to promote potential probiotics, therefore controlling inflammation in AS.
Collapse
Affiliation(s)
- Yupeng Lai
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Wenli Tang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao Luo
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Huihui Zheng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yanpeng Zhang
- Department of Laboratory, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Meiying Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Guangchuang Yu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Min Yang
- Department of Rheumatology and Immunology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Li W, Jiang J, Yin X, Zhang Y, Zou X, Sun M, Jia J, Ma B, Xu J. Mediation of Regional Cerebral Blood Flow in the Relationship between Specific Gut Microbiota and Cognition in Vascular Cognitive Impairment. J Alzheimers Dis 2024; 97:435-445. [PMID: 38108351 DOI: 10.3233/jad-230709] [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] [Indexed: 12/19/2023]
Abstract
BACKGROUND Gut microbiota could affect the onset and development of vascular cognitive impairment (VCI) through modulating metabolic and immune pathways. However, the vascular mechanisms involved remain unclear. OBJECTIVE To investigate the gut microbiota associated with VCI and examine the mediating effects of regional cerebral blood flow (CBF) to explore potential therapeutic targets for VCI. METHODS This prospective study enrolled patients with VCI (n = 16) and healthy controls (n = 18) from the Chinese Imaging, Biomarkers, and Lifestyle study between January 1 and June 30, 2022. The gut microbiota composition and diversity were determined by 16 S ribosomal RNA gene sequencing. The association between gut microbiota and Montreal Cognitive Assessment (MoCA) scores was determined using Spearman's correlation analysis. Regional CBF was calculated using pseudo-continuous arterial spin labeling. The mediating effects of regional CBF on the relationship between specific gut microbiota and cognition in VCI were investigated using mediation analysis. RESULTS Compared to healthy controls, patients with VCI had significantly greater abundance of Bifidobacterium, Veillonella, R uminococcus gnavus , Fusobacterium, and Erysipelatoclostridium and smaller abundance of Collinsella. The abundance of Ruminococcus gnavus was negatively associated with MoCA scores in patients with VCI, with the CBF in the left hypothalamus, right hypothalamus, and left amygdala accounting for 63.96%, 48.22%, and 36.51%, respectively, of this association after adjusting for confounders. CONCLUSIONS Ruminococcus gnavus is associated with cognition in VCI, which is strongly mediated by CBF in the bilateral hypothalamus and left amygdala. These findings highlight the potential regulatory roles of nutrition and metabolism-related areas of the brain in VCI.
Collapse
Affiliation(s)
- Wenyi Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jiwei Jiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | | | - Yuan Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xinying Zou
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Mengfan Sun
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jianjun Jia
- Department of Geriatric Neurology, The Second Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Baiping Ma
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Jun Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
14
|
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: 1] [Impact Index Per Article: 0.5] [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
|
15
|
Juge N. Microbe Profile: Ruminococcus gnavus: the yin and yang of human gut symbionts. MICROBIOLOGY (READING, ENGLAND) 2023; 169. [PMID: 37622435 PMCID: PMC10482369 DOI: 10.1099/mic.0.001383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/03/2023] [Indexed: 08/26/2023]
Abstract
Graphical abstract
Collapse
Affiliation(s)
- Nathalie Juge
- Quadram Institute Bioscience, Food, Microbiome and Health Institute Strategic Programme, Norwich NR4 7UQ, UK
| |
Collapse
|
16
|
Thompson KN, Bonham KS, Ilott NE, Britton GJ, Colmenero P, Bullers SJ, McIver LJ, Ma S, Nguyen LH, Filer A, Brough I, Pearson C, Moussa C, Kumar V, Lam LH, Jackson MA, Pawluk A, Kiriakidis S, Taylor PC, Wedderburn LR, Marsden B, Young SP, Littman DR, Faith JJ, Pratt AG, Bowness P, Raza K, Powrie F, Huttenhower C. Alterations in the gut microbiome implicate key taxa and metabolic pathways across inflammatory arthritis phenotypes. Sci Transl Med 2023; 15:eabn4722. [PMID: 37494472 DOI: 10.1126/scitranslmed.abn4722] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 06/22/2023] [Indexed: 07/28/2023]
Abstract
Musculoskeletal diseases affect up to 20% of adults worldwide. The gut microbiome has been implicated in inflammatory conditions, but large-scale metagenomic evaluations have not yet traced the routes by which immunity in the gut affects inflammatory arthritis. To characterize the community structure and associated functional processes driving gut microbial involvement in arthritis, the Inflammatory Arthritis Microbiome Consortium investigated 440 stool shotgun metagenomes comprising 221 adults diagnosed with rheumatoid arthritis, ankylosing spondylitis, or psoriatic arthritis and 219 healthy controls and individuals with joint pain without an underlying inflammatory cause. Diagnosis explained about 2% of gut taxonomic variability, which is comparable in magnitude to inflammatory bowel disease. We identified several candidate microbes with differential carriage patterns in patients with elevated blood markers for inflammation. Our results confirm and extend previous findings of increased carriage of typically oral and inflammatory taxa and decreased abundance and prevalence of typical gut clades, indicating that distal inflammatory conditions, as well as local conditions, correspond to alterations to the gut microbial composition. We identified several differentially encoded pathways in the gut microbiome of patients with inflammatory arthritis, including changes in vitamin B salvage and biosynthesis and enrichment of iron sequestration. Although several of these changes characteristic of inflammation could have causal roles, we hypothesize that they are mainly positive feedback responses to changes in host physiology and immune homeostasis. By connecting taxonomic alternations to functional alterations, this work expands our understanding of the shifts in the gut ecosystem that occur in response to systemic inflammation during arthritis.
Collapse
Affiliation(s)
- Kelsey N Thompson
- Department of Biostatistics, T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Kevin S Bonham
- Department of Biostatistics, T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nicholas E Ilott
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedic, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford OX3 7FY, UK
| | - Graham J Britton
- Marc and Jennifer Lipschultz Precision Immunology Institute and Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paula Colmenero
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedic, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford OX3 7FY, UK
| | - Samuel J Bullers
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedic, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford OX3 7FY, UK
| | - Lauren J McIver
- Department of Biostatistics, T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Siyuan Ma
- Department of Biostatistics, T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Long H Nguyen
- Department of Biostatistics, T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Andrew Filer
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham B15 2TT, UK
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research and Research Into Inflammatory Arthritis Centre Versus Arthritis, University of Birmingham, Chesterfield S41 7TD, UK
| | - India Brough
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedic, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford OX3 7FY, UK
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Claire Pearson
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedic, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford OX3 7FY, UK
| | - Caroline Moussa
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedic, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford OX3 7FY, UK
| | - Vinod Kumar
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedic, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford OX3 7FY, UK
| | - Lilian H Lam
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedic, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford OX3 7FY, UK
| | - Matthew A Jackson
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedic, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford OX3 7FY, UK
| | - April Pawluk
- Department of Biostatistics, T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Serafim Kiriakidis
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Peter C Taylor
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Lucy R Wedderburn
- Centre for Adolescent Rheumatology Versus Arthritis, University College London, UCLH, and GOSH, Chesterfield S41 7TD, UK
- NIHR Great Ormond Street Biomedical Research Centre, University College London, London WC1N 1EH, UK
- UCL GOS Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Brian Marsden
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedic, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford OX3 7FY, UK
| | - Stephen P Young
- Department of Rheumatology, Sandwell & West Birmingham NHS Trust, West Bromwich B71 4HJ, UK
| | - Dan R Littman
- Howard Hughes Medical Institute and the Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Jeremiah J Faith
- Marc and Jennifer Lipschultz Precision Immunology Institute and Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Arthur G Pratt
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Research into Inflammatory Arthritis Centre Versus Arthritis, Newcastle Birmingham, Glasgow, and Oxford, Chesterfield S41 7TD, UK
- Department of Rheumatology, Musculoskeletal Services Directorate, Newcastle upon Tyne Hospitals NHS Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Paul Bowness
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Karim Raza
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham B15 2TT, UK
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research and Research Into Inflammatory Arthritis Centre Versus Arthritis, University of Birmingham, Chesterfield S41 7TD, UK
- Department of Rheumatology, Sandwell & West Birmingham NHS Trust, West Bromwich B71 4HJ, UK
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedic, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford OX3 7FY, UK
| | - Curtis Huttenhower
- Department of Biostatistics, T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
17
|
Fan KC, Lin CC, Liu YC, Chao YP, Lai YJ, Chiu YL, Chuang YF. Altered gut microbiota in older adults with mild cognitive impairment: a case-control study. Front Aging Neurosci 2023; 15:1162057. [PMID: 37346147 PMCID: PMC10281289 DOI: 10.3389/fnagi.2023.1162057] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/02/2023] [Indexed: 06/23/2023] Open
Abstract
Introduction The microbiota-gut-brain axis is implicated in Alzheimer's disease. Gut microbiota alterations in mild cognitive impairment (MCI) are inconsistent and remain to be understood. This study aims to investigate the gut microbial composition associated with MCI, cognitive functions, and structural brain differences. Methods A nested case-control study was conducted in a community-based prospective cohort where detailed cognitive functions and structural brain images were collected. Thirty-one individuals with MCI were matched to sixty-five cognitively normal controls by age strata, gender, and urban/rural area. Fecal samples were examined using 16S ribosomal RNA (rRNA) V3-V4 sequencing. Compositional differences between the two groups were identified and correlated with the cognitive functions and volumes/thickness of brain structures. Results There was no significant difference in alpha and beta diversity between MCIs and cognitively normal older adults. However, the abundance of the genus Ruminococcus, Butyricimonas, and Oxalobacter decreased in MCI patients, while an increased abundance of nine other genera, such as Flavonifractor, were found in MCIs. Altered genera discriminated MCI patients well from controls (AUC = 84.0%) and were associated with attention and executive function. Conclusion This study provides insights into the role of gut microbiota in the neurodegenerative process.
Collapse
Affiliation(s)
- Kang-Chen Fan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chen-Ching Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Chien Liu
- Department of Neurology, Cardinal Tien Hospital, New Taipei, Taiwan
| | - Yi-Ping Chao
- Department of Computer Science and Information Engineering, Chang Gung University, Taoyuan, Taiwan
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yen-Jun Lai
- Division of Medical Imaging, Department of Radiology, Far Eastern Memorial Hospital, New Taipei, Taiwan
| | - Yen-Ling Chiu
- Department of Medical Research, Far Eastern Memorial Hospital, Taipei, Taiwan
- Graduate Program in Biomedical Informatics and Graduate Institute of Medicine, Yuan Ze University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Fang Chuang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Public Health, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Psychiatry, Far Eastern Memorial Hospital, New Taipei, Taiwan
| |
Collapse
|
18
|
Meng S, Zhang Y, Lv S, Zhang Z, Liu X, Jiang L. Comparison of muscle metabolomics between two Chinese horse breeds. Front Vet Sci 2023; 10:1162953. [PMID: 37215482 PMCID: PMC10196265 DOI: 10.3389/fvets.2023.1162953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/03/2023] [Indexed: 05/24/2023] Open
Abstract
With their enormous muscle mass and athletic ability, horses are well-positioned as model organisms for understanding muscle metabolism. There are two different types of horse breeds-Guanzhong (GZ) horses, an athletic breed with a larger body height (~148.7 cm), and the Ningqiang pony (NQ) horses, a lower height breed generally used for ornamental purposes-both inhabited in the same region of China with obvious differences in muscle content. The main objective of this study was to evaluate the breed-specific mechanisms controlling muscle metabolism. In this study, we observed muscle glycogen, enzyme activities, and LC-MS/MS untargeted metabolomics in the gluteus medius muscle of six, each of GZ and NQ horses, to explore differentiated metabolites that are related to the development of two muscles. As expected, the glycogen content, citrate synthase, and hexokinase activity of muscle were significantly higher in GZ horses. To alleviate the false positive rate, we used both MS1 and MS2 ions for metabolite classification and differential analysis. As a result, a total of 51,535 MS1 and 541 MS2 metabolites were identified, and these metabolites can separate these two groups from each other. Notably, 40% of these metabolites were clustered into lipids and lipid-like molecules. Furthermore, 13 significant metabolites were differentially detected between GZ and NQ horses (fold change [FC] value ≥ 2, variable important in projection value ≥1, and Q value ≤ 0.05). They are primarily clustered into glutathione metabolism (GSH, p = 0.01), taurine, and hypotaurine metabolism (p < 0.05) pathways. Seven of the 13 metabolites were also found in thoroughbred racing horses, suggesting that metabolites related to antioxidants, amino acids, and lipids played a key role in the development of skeleton muscle in horses. Those metabolites related to muscle development shed a light on racing horses' routine maintenance and improvement of athletic performance.
Collapse
Affiliation(s)
- Sihan Meng
- Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Yanli Zhang
- Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Shipeng Lv
- College of Animal Science, Xinjiang Agricultural University, Urumqi, China
| | - Zhengkai Zhang
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Xuexue Liu
- Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
- Centre d'Anthropobiologie et de Génomique de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Lin Jiang
- Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| |
Collapse
|
19
|
Crost EH, Coletto E, Bell A, Juge N. Ruminococcus gnavus: friend or foe for human health. FEMS Microbiol Rev 2023; 47:fuad014. [PMID: 37015876 PMCID: PMC10112845 DOI: 10.1093/femsre/fuad014] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 02/06/2023] [Accepted: 04/03/2023] [Indexed: 04/06/2023] Open
Abstract
Ruminococcus gnavus was first identified in 1974 as a strict anaerobe in the gut of healthy individuals, and for several decades, its study has been limited to specific enzymes or bacteriocins. With the advent of metagenomics, R. gnavus has been associated both positively and negatively with an increasing number of intestinal and extraintestinal diseases from inflammatory bowel diseases to neurological disorders. This prompted renewed interest in understanding the adaptation mechanisms of R. gnavus to the gut, and the molecular mediators affecting its association with health and disease. From ca. 250 publications citing R. gnavus since 1990, 94% were published in the last 10 years. In this review, we describe the biological characterization of R. gnavus, its occurrence in the infant and adult gut microbiota and the factors influencing its colonization of the gastrointestinal tract; we also discuss the current state of our knowledge on its role in host health and disease. We highlight gaps in knowledge and discuss the hypothesis that differential health outcomes associated with R. gnavus in the gut are strain and niche specific.
Collapse
Affiliation(s)
- Emmanuelle H Crost
- Quadram Institute Bioscience, Rosalind Franklin Road, Colney, Norwich NR4 7UQ, United Kingdom
| | - Erika Coletto
- Quadram Institute Bioscience, Rosalind Franklin Road, Colney, Norwich NR4 7UQ, United Kingdom
| | - Andrew Bell
- Quadram Institute Bioscience, Rosalind Franklin Road, Colney, Norwich NR4 7UQ, United Kingdom
| | - Nathalie Juge
- Quadram Institute Bioscience, Rosalind Franklin Road, Colney, Norwich NR4 7UQ, United Kingdom
| |
Collapse
|
20
|
Bell A, Severi E, Owen CD, Latousakis D, Juge N. Biochemical and structural basis of sialic acid utilization by gut microbes. J Biol Chem 2023; 299:102989. [PMID: 36758803 PMCID: PMC10017367 DOI: 10.1016/j.jbc.2023.102989] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
The human gastrointestinal (GI) tract harbors diverse microbial communities collectively known as the gut microbiota that exert a profound impact on human health and disease. The repartition and availability of sialic acid derivatives in the gut have a significant impact on the modulation of gut microbes and host susceptibility to infection and inflammation. Although N-acetylneuraminic acid (Neu5Ac) is the main form of sialic acids in humans, the sialic acid family regroups more than 50 structurally and chemically distinct modified derivatives. In the GI tract, sialic acids are found in the terminal location of mucin glycan chains constituting the mucus layer and also come from human milk oligosaccharides in the infant gut or from meat-based foods in adults. The repartition of sialic acid in the GI tract influences the gut microbiota composition and pathogen colonization. In this review, we provide an update on the mechanisms underpinning sialic acid utilization by gut microbes, focusing on sialidases, transporters, and metabolic enzymes.
Collapse
Affiliation(s)
- Andrew Bell
- Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich, United Kingdom
| | - Emmanuele Severi
- Microbes in Health and Disease, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - C David Owen
- Diamond Light Source Ltd, Diamond House, Harwell Science and Innovation Campus, Didcot, United Kingdom
| | - Dimitrios Latousakis
- Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich, United Kingdom
| | - Nathalie Juge
- Quadram Institute Bioscience, Gut Microbes and Health Institute Strategic Programme, Norwich, United Kingdom.
| |
Collapse
|
21
|
Xu H, Wang S, Jiang Y, Wu J, Chen L, Ding Y, Zhou Y, Deng L, Chen X. Poria cocos Polysaccharide Ameliorated Antibiotic-Associated Diarrhea in Mice via Regulating the Homeostasis of the Gut Microbiota and Intestinal Mucosal Barrier. Int J Mol Sci 2023; 24:1423. [PMID: 36674937 PMCID: PMC9862632 DOI: 10.3390/ijms24021423] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
Poria cocos polysaccharides (PCP) have been validated for several biological activities, including antitumor, anti-inflammatory, antioxidant, immunomodulatory, hepatoprotective and modulation on gut microbiota. In this research, we aim to demonstrate the potential prebiotic effects and the therapeutic efficacies of PCP in the treatment of antibiotic-associated diarrhea (AAD), and confirm the beneficial effects of PCP on gut dysbiosis. Antibiotic-associated diarrhea mice models were established by treating them with broad-spectrum antibiotics in drinking water for seven days. Mice in two groups treated with probiotics and polysaccharide were given Bifico capsules (4.2 g/kg/d) and PCP (250 mg/kg/d) for seven days using intragastric gavage, respectively. To observe the regulatory effects of PCP on gut microbiota and intestinal mucosal barrier, we conducted the following experiments: intestinal flora analysis (16S rDNA sequencing), histology (H&E staining) and tight junction proteins (immunofluorescence staining). The levels of mRNA expression of receptors associated with inflammation and gut metabolism were assessed by real-time reverse transcription-polymerase chain reaction (RT-PCR). The study revealed that PCP can comprehensively improve the clinical symptoms of AAD mice, including fecal traits, mental state, hair quality, etc., similar to the effect of probiotics. Based on histology observation, PCP significantly improved the substantial structure of the intestine of AAD mice by increasing the expression levels of colonic tight junction protein zonula-occludens 1 (ZO-1) and its mRNA. Moreover, PCP not only increased the abundance of gut microbiota, but also increased the diversity of gut microbiota in AAD mice, including alpha diversity and beta diversity. Further analysis found that PCP can modulate seven characteristic species of intestinal flora in AAD mice, including Parabacteroides_distasonis, Akkermansia_muciniphila, Clostridium_saccharolyticum, Ruminoc-occus_gnavus, Lactobacillus_salivarius, Salmonella_enterica and Mucispirillum_schaedleri. Finally, enrichment analysis predicted that PCP may affect intestinal mucosal barrier function, host immune response and metabolic function by regulating the microbiota. RT-PCR experiments showed that PCP can participate in immunomodulatory and modulation on metabolic by regulating the mRNA expression of forkhead-box protein 3 (FOXP3) and G protein-coupled receptor 41 (GPR41). These results indicated that Poria cocos polysaccharide may ameliorate antibiotic-associated diarrhea in mice by regulating the homeostasis of the gut microbiota and intestinal mucosal barrier. In addition, polysaccharide-derived changes in intestinal microbiota were involved in the immunomodulatory activities and modulation of the metabolism.
Collapse
Affiliation(s)
- Huachong Xu
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Guangzhou 510632, China
| | - Shiqi Wang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Yawen Jiang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Jialin Wu
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Lili Chen
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Yujia Ding
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Yingtong Zhou
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Li Deng
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Guangzhou 510632, China
| | - Xiaoyin Chen
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Guangzhou 510632, China
| |
Collapse
|
22
|
Yuan L, Zhang R, Li X, Gao C, Hu X, Hussain S, Zhang L, Wang M, Ma X, Pan Q, Lou X, Si S. Long-term simulated microgravity alters gut microbiota and metabolome in mice. Front Microbiol 2023; 14:1100747. [PMID: 37032862 PMCID: PMC10080065 DOI: 10.3389/fmicb.2023.1100747] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Abstract
Spaceflight and microgravity has a significant impact on the immune, central nervous, bone, and muscle support and cardiovascular systems. However, limited studies are available on the adverse effects of long-term microgravity on the intestinal microbiota, metabolism, and its relationships. In this study, a ground-based simulated microgravity (SMG) mouse model was established to evaluate the impact of long-term microgravity on gut microbiota and metabolome. After 8 weeks of SMG, alterations of the intestinal microbiota and metabolites were detected using 16S rRNA sequencing and untargeted metabolomics. Compared to the control, no significant differences in α-diversity were observed at weeks 2, 4 and 8. Nevertheless, there were clear differences in community structures at different time points. The phylum Verrucomicrobia significantly declined from 2 to 8 weeks of SMG, yet the relative abundance of Actinobacteria and Deferribacteres expanded remarkably at weeks 8. SMG decreased the genus of Allobaculum and increased Bacteroides significantly throughout the period of 8 weeks. Besides, Genus Akkermansia, Gracilibacter, Prevotella, Odoribacter, Rothia, Sporosarcina, Gracilibacter, Clostridium, and Mucispirillum were identified as biomarkers for SMG group. Desulfovibrio_c21_c20, Akkermansia_muciniphila, and Ruminococcus_gnavus dropped at week 2, which tend to recover at week 4, except for Akkermansia_muciniphila. Bacteroides_uniformis and Faecalibacterium_prausnitzii declined significantly, while Ruminococcus_flavefaciens and Mucispirillum_schaedleri elevated at week 8. Furthermore, intestinal metabolome analysis showed that 129 were upregulated and 146 metabolites were downregulated in SMG. Long-term SMG most affected steroid hormone biosynthesis, tryptophan, cysteine, methionine, arginine, proline metabolism, and histidine metabolism. Correlated analysis suggested that the potential beneficial taxa Allobaculum, Akkermansia, and Faecalibacterium were negatively associated with tryptophan, histidine, arginine, and proline metabolism, but positively with steroid hormone biosynthesis. Yet Bacteroides, Lachnospiraceae_Clostridium, Rothia, Bilophila, and Coprococcus were positively correlated with arginine, proline, tryptophan, and histidine metabolism, while negatively associated with steroid hormone biosynthesis. These results suggest that Long-term SMG altered the community of intestinal microbiota, and then further disturbed intestinal metabolites and metabolic pathways, which have great potential to help understand and provide clues for revealing the mechanisms of long-term SMG involved diseases.
Collapse
Affiliation(s)
- Lu Yuan
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Rong Zhang
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Xinlou Li
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Caiyun Gao
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Xiangnan Hu
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Safdar Hussain
- Center for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Linlin Zhang
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Moye Wang
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Xiaoyu Ma
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
| | - Qiuxia Pan
- Department of Traditional Chinese Medicine, PLA Strategic Support Force Medical Center, Beijing, China
| | - Xiaotong Lou
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
- *Correspondence: Xiaotong Lou,
| | - Shaoyan Si
- Department of Medical Research, PLA Strategic Support Force Medical Center, Beijing, China
- Shaoyan Si,
| |
Collapse
|
23
|
Chen Z, Wang Z, Li D, Zhu B, Xia Y, Wang G, Ai L, Zhang C, Wang C. The gut microbiota as a target to improve health conditions in a confined environment. Front Microbiol 2022; 13:1067756. [PMID: 36601399 PMCID: PMC9806127 DOI: 10.3389/fmicb.2022.1067756] [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: 10/12/2022] [Accepted: 11/22/2022] [Indexed: 12/23/2022] Open
Abstract
Confined environments increase psychological stress and lead to health problems such as abnormal mood and rhythm disruption. However, the mechanism by which confined environments impact health has remained unclear. Significant correlations have been reported between psychological stress and changes in gut microbiota. Therefore, we investigated the effect of a confined environment on the composition of the gut microbiota by 16s rDNA high-throughput sequencing, and analyzed the correlation between gut microbiota and health indicators such as uric acid (UA), sleep, and mood. We found that the gut microbiota of the subjects clustered into two enterotypes (Bi and Bla), and that the groups differed significantly. There were notable differences in the abundances of genera such as Bifidobacterium, Dorea, Ruminococcus_torques_group, Ruminococcus_gnavus_group, Klebsiella, and UCG-002 (p < 0.05). A confined environment significantly impacted the subjects' health indicators. We also observed differences in how the subjects of the two enterotypes adapted to the confined environment. The Bi group showed no significant differences in health indicators before and after confinement; however, the Bla group experienced several health problems after confinement, such as increased UA, anxiety, and constipation, and lack of sleep. Redundancy analysis (RDA) showed that UA, RBC, mood, and other health problems were significantly correlated with the structure of the gut microbiota. We concluded that genera such as UCG-002, Ruminococcus, CAG352, and Ruminococcus_torques_group increased vulnerability to confined environments, resulting in abnormal health conditions. We found that the differences in the adaptability of individuals to confined environments were closely related to the composition of their gut microbiota.
Collapse
Affiliation(s)
- Zheng Chen
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - ZiYing Wang
- Navy Special Medical Center, Naval Medical University, Shanghai, China
| | - Dan Li
- Navy Special Medical Center, Naval Medical University, Shanghai, China
| | - Beiwei Zhu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Yongjun Xia
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Guangqiang Wang
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Lianzhong Ai
- School of Health Science and Engineering, Shanghai Engineering Research Center of Food Microbiology, University of Shanghai for Science and Technology, Shanghai, China
| | - Chunhong Zhang
- Navy Special Medical Center, Naval Medical University, Shanghai, China,*Correspondence: Chunhong Zhang,
| | - Chuan Wang
- Navy Special Medical Center, Naval Medical University, Shanghai, China,Chuan Wang,
| |
Collapse
|