1
|
Laplanche V, Armiento S, Speciale I, Šuligoj T, Crost EH, Lamprinaki D, Vaux L, Gotts K, De Castro C, Juge N. The human gut symbiont Ruminococcus gnavus displays strain-specific exopolysaccharides modulating the host immune response. Carbohydr Polym 2025; 347:122754. [PMID: 39486983 DOI: 10.1016/j.carbpol.2024.122754] [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: 06/02/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 11/04/2024]
Abstract
Ruminococcus gnavus is a prevalent member of the human gut microbiota and over-represented in inflammatory bowel diseases. R. gnavus ATCC 29149 was previously shown to produce a pro-inflammatory exopolysaccharide (EPS) referred to here as glucorhamnan-I or EPS29149. Here, we determined the structure of the polysaccharides from R. gnavus ATCC 35913 (EPS35193) and E1 (EPSE1) strains, both consist of a repeating unit with a backbone composed of four α-L-rhamnose units, with alternate 2- and 3-linkages, and a β-d-glucose residue linked to O-2 of one 3-Rha as side branch. This structure differs from EPS29149 and is referred to as glucorhamnan-II. EPS35193 and EPSE1 showed variation in the glucosylation level that is non-stochiometric in EPS35193.R. gnavus strains and their purified EPS induced strain-specific production of cytokines and chemokines in bone-marrow derived dendritic cells and NF-κB activation in reporter cells. R. gnavus ATCC 35913 was the most immunogenic strain, likely due to the absence of an additional capsular polysaccharide layer as shown by TEM, while EPS29149, EPS35193 and EPSE1 showed activation of TLR4 reporter cells. These strain-specific differences in R. gnavus cell surface glycosylation and host response underscore the importance of studying R. gnavus-host interaction at the strain level.
Collapse
Affiliation(s)
- Victor Laplanche
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Samantha Armiento
- Department of Chemical Sciences, University of Naples Federico II, via Cintia, 26, 80126 Napoli, Italy
| | - Immacolata Speciale
- Department of Chemical Sciences, University of Naples Federico II, via Cintia, 26, 80126 Napoli, Italy
| | - Tanja Šuligoj
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Emmanuelle H Crost
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Dimitra Lamprinaki
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Laura Vaux
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Kathryn Gotts
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK
| | - Cristina De Castro
- Department of Chemical Sciences, University of Naples Federico II, via Cintia, 26, 80126 Napoli, Italy.
| | - Nathalie Juge
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, UK.
| |
Collapse
|
2
|
Valentino V, De Filippis F, Marotta R, Pasolli E, Ercolini D. Genomic features and prevalence of Ruminococcus species in humans are associated with age, lifestyle, and disease. Cell Rep 2024; 43:115018. [PMID: 39615045 DOI: 10.1016/j.celrep.2024.115018] [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: 05/15/2024] [Revised: 09/23/2024] [Accepted: 11/11/2024] [Indexed: 12/28/2024] Open
Abstract
The genus Ruminococcus is dominant in the human gut, but higher levels of some species, such as R. gnavus, R. torques, and R. bromii, have been linked to health or disease. In this study, we analyzed >9,000 Ruminococcus metagenome-assembled genomes (MAGs) reconstructed from >5,000 subjects and revealed significant links between the prevalence of some species/subspecies and geographic origin, age, lifestyle, and disease, with subspecies prevalent in specific subpopulations showing divergent metabolic potential. Furthermore, Ruminococcus species from Lachnospiraceae encoded for carbohydrate-active enzymes (CAZy) potentially involved in the metabolism of human N- and O-glycans, whereas those from Oscillospiraceae appear to be more adapted toward fiber metabolism. These new findings contribute to elucidating the potential functional role of Ruminococcus in specific lifestyles and diseases and to decipher the diversity and the adaptation of members of this genus to the human gut.
Collapse
Affiliation(s)
- Vincenzo Valentino
- Department of Agricultural Sciences, University of Naples Federico II, Piazza Carlo di Borbone 1, Portici, 80055 Naples, Italy
| | - Francesca De Filippis
- Department of Agricultural Sciences, University of Naples Federico II, Piazza Carlo di Borbone 1, Portici, 80055 Naples, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Roberto Marotta
- Department of Agricultural Sciences, University of Naples Federico II, Piazza Carlo di Borbone 1, Portici, 80055 Naples, Italy
| | - Edoardo Pasolli
- Department of Agricultural Sciences, University of Naples Federico II, Piazza Carlo di Borbone 1, Portici, 80055 Naples, Italy
| | - Danilo Ercolini
- Department of Agricultural Sciences, University of Naples Federico II, Piazza Carlo di Borbone 1, Portici, 80055 Naples, Italy; Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
Han H, Gao M, Wang F, Luo Z, Jiang X, Qiu Y, Su J, Duan X, Luo S, Tang S, Khan A, Zou Z, Chen C, Yin Q, Qiu J, Zhang H. Protective effects of patchouli alcohol against DSS-induced ulcerative colitis. Sci Rep 2024; 14:16745. [PMID: 39033185 PMCID: PMC11271309 DOI: 10.1038/s41598-024-66259-8] [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: 02/22/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024] Open
Abstract
Patchouli alcohol (PA) is a widely used pharmaceutical ingredient in various Chinese traditional herbal medicine (THM) formulations, known for its modulatory effects on the gut microbiota. The present study investigated PA's anti-inflammatory and regulatory effects on gut microbiota and its mode of action (MOA). Based on the assessments of ulcerative colitis (UC) symptoms, PA exhibited promising preventions against inflammatory response. In accordance, the expressions of pro-inflammatory factors, including interleukin (IL)-1β, IL-6, tumor necrosis factor-α, and chemokine ligand 5 were significantly attenuated under PA treatment. Furthermore, PA enhanced the intestinal barrier damage caused by dextran sodium sulfate (DSS). Interestingly, PA exhibited negligible inventions on DSS-induced gut microbiota dysbiosis. PA did not affect the diversity of the DSS gut microbiota, it did alter the composition, as evidenced by a significant increase in the Firmicutes-Bacteroidetes (F/B) ratio. Finally, the MOA of PA against inflammation in DSS-treated mice was addressed by suppressing the expressions of heme oxygenase-1 (HO-1) and inducible nitric oxide synthase (iNOS). In conclusion, PA prevented inflammatory response in the DSS-induced UC mice model via directly suppressing HO-1 and iNOS-associated antioxidant signal pathways, independent of its effects on gut microbiota composition.
Collapse
Affiliation(s)
- Huifang Han
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Min Gao
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Fanghong Wang
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Zheng Luo
- Jiulongpo District Center for Disease Control and Prevention, Chongqing Municipality, Chongqing, 400039, People's Republic of China
| | - Xuejun Jiang
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yu Qiu
- Department of Neurology, The Affiliated University-Town Hospital of Chongqing Medical University, Chongqing, 401331, People's Republic of China
| | - Junhao Su
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Xinhao Duan
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Shiyue Luo
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Shixin Tang
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China
| | - Ahmad Khan
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Zhen Zou
- Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Qi Yin
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China.
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Jingfu Qiu
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China.
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Hongyang Zhang
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, No. 1, Yixueyuan Road, Yuzhong District, Chongqing, 400016, People's Republic of China.
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
6
|
Luo P, Gao D, Zhang Q. Genetic causal relationship between gut microbiota and basal cell carcinoma: A two-sample mendelian randomization study. Skin Res Technol 2024; 30:e13804. [PMID: 38895789 PMCID: PMC11187847 DOI: 10.1111/srt.13804] [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: 05/14/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVE Research has previously established connections between the intestinal microbiome and the progression of some cancers. However, there is a noticeable gap in the literature in regard to using Mendelian randomisation (MR) to delve into potential causal relationships between the gut microbiota (GM) and basal cell carcinoma (BCC). Therefore, the purpose of our study was to use MR to explore the causal relationship between four kinds of GM (Bacteroides, Streptococcus, Proteobacteria and Lachnospiraceae) and BCC. METHODS We used genome-wide association study (GWAS) data and MR to explore the causal relationship between four kinds of GM and BCC. This study primarily employed the random effect inverse variance weighted (IVW) model for analysis, as complemented by additional methods including the simple mode, weighted median, weighted mode and MR‒Egger methods. We used heterogeneity and horizontal multiplicity to judge the reliability of each analysis. MR-PRESSO was mainly used to detect and correct outliers. RESULTS The random-effects IVW results showed that Bacteroides (OR = 0.936, 95% CI = 0.787-1.113, p = 0.455), Streptococcus (OR = 0.974, 95% CI = 0.875-1.083, p = 0.629), Proteobacteria (OR = 1.113, 95% CI = 0.977-1.267, p = 0.106) and Lachnospiraceae (OR = 1.027, 95% CI = 0.899-1.173, p = 0.688) had no genetic causal relationship with BCC. All analyses revealed no horizontal pleiotropy, heterogeneity or outliers. CONCLUSION We found that Bacteroides, Streptococcus, Proteobacteria and Lachnospiraceae do not increase the incidence of BCC at the genetic level, which provides new insight for the study of GM and BCC.
Collapse
Affiliation(s)
- Pan Luo
- Department of Comprehensive Plastic SurgeryPlastic Surgery HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Dejin Gao
- Department of Comprehensive Plastic SurgeryPlastic Surgery HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qingguo Zhang
- Department of Comprehensive Plastic SurgeryPlastic Surgery HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
7
|
Fu J, Hao Z. The causality between gut microbiota and non-Hodgkin lymphoma: a two-sample bidirectional Mendelian randomization study. Front Microbiol 2024; 15:1403825. [PMID: 38860220 PMCID: PMC11163074 DOI: 10.3389/fmicb.2024.1403825] [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: 03/20/2024] [Accepted: 05/10/2024] [Indexed: 06/12/2024] Open
Abstract
Background Studies have indicated an association between gut microbiota (GM) and non-Hodgkin lymphoma (NHL). However, the causality between GM and NHL remains unclear. This study aims to investigate the causality between GM and NHL using Mendelian randomization (MR). Methods Data on GM is sourced from the MiBioGen consortium, while data on NHL and its subtypes is sourced from the FinnGen consortium R10 version. Inverse variance weighted (IVW) was employed for the primary MR analysis method, with methods such as Bayesian weighted Mendelian randomisation (BWMR) as an adjunct. Sensitivity analyses were conducted using Cochran's Q test, MR-Egger regression, MR-PRESSO, and the "Leave-one-out" method. Results The MR results showed that there is a causality between 27 GMs and NHL. Among them, 20 were negatively associated (OR < 1), and 7 were positively associated (OR > 1) with the corresponding diseases. All 27 MR results passed sensitivity tests, and there was no reverse causal association. Conclusion By demonstrating a causal link between GM and NHL, this research offers novel ideas to prevent, monitor, and cure NHL later.
Collapse
Affiliation(s)
- Jinjie Fu
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zheng Hao
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin, China
- Guo Aichun Institute of Medical History and Literature, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
8
|
Zhang M, Shi Z, Wu C, Yang F, Su T, Jing X, Shi J, Ren H, Jiang L, Jiang Y, Zhang C, Zhou W, Zhou Y, Wu K, Zheng S, Zhong X, Wu L, Gu W, Hong J, Wang J, Ning G, Liu R, Zhong H, Zhou W, Wang W. Cushing Syndrome Is Associated With Gut Microbial Dysbiosis and Cortisol-Degrading Bacteria. J Clin Endocrinol Metab 2024; 109:1474-1484. [PMID: 38157274 DOI: 10.1210/clinem/dgad766] [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: 08/29/2023] [Revised: 11/28/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024]
Abstract
CONTEXT Cushing syndrome (CS) is a severe endocrine disease characterized by excessive secretion of cortisol with multiple metabolic disorders. While gut microbial dysbiosis plays a vital role in metabolic disorders, the role of gut microbiota in CS remains unclear. OBJECTIVE The objective of this work is to examine the alteration of gut microbiota in patients with CS. METHODS We performed shotgun metagenomic sequencing of fecal samples from 78 patients with CS and 78 healthy controls matched for age and body mass index. Furthermore, we verify the cortisol degradation capacity of Ruminococcus gnavus in vitro and identify the potential metabolite by LC-MC/MS. RESULTS We observed significant differences in microbial composition between CS and controls in both sexes, with CS showing reduced Bacteroidetes (Bacteroides vulgatus) and elevated Firmicutes (Erysipelotrichaceae_bacterium_6_1_45) and Proteobacteria (Enterobacter cloacae). Despite distinct causes of hypercortisolism in ACTH-dependent and ACTH-independent CS, we found no significant differences in metabolic profiles or gut microbiota between the 2 subgroups. Furthermore, we identified a group of gut species, including R. gnavus, that were positively correlated with cortisol levels in CS. These bacteria were found to harbor cortisol-degrading desAB genes and were consistently enriched in CS. Moreover, we demonstrated the efficient capacity of R. gnavus to degrade cortisol to 11-oxygenated androgens in vitro. CONCLUSION This study provides evidence of gut microbial dysbiosis in patients with CS and identifies a group of CS-enriched bacteria capable of degrading cortisol. These findings highlight the potential role of gut microbiota in regulating host steroid hormone levels, and consequently host health.
Collapse
Affiliation(s)
- Minchun Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhun Shi
- BGI Research, Shenzhen 518083, China
| | - Chao Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | | | - Tingwei Su
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaohuan Jing
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Juan Shi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | | | - Lei Jiang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yiran Jiang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Cui Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenzhong Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yijing Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Kui Wu
- BGI Research, Shenzhen 518083, China
| | - Sichang Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xu Zhong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Luming Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | | | - Weiwei Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
9
|
Lin X, Hu T, Wu Z, Li L, Wang Y, Wen D, Liu X, Li W, Liang H, Jin X, Xu X, Wang J, Yang H, Kristiansen K, Xiao L, Zou Y. Isolation of potentially novel species expands the genomic and functional diversity of Lachnospiraceae. IMETA 2024; 3:e174. [PMID: 38882499 PMCID: PMC11170972 DOI: 10.1002/imt2.174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/06/2023] [Indexed: 06/18/2024]
Abstract
The Lachnospiraceae family holds promise as a source of next-generation probiotics, yet a comprehensive delineation of its diversity is lacking, hampering the identification of suitable strains for future applications. To address this knowledge gap, we conducted an in-depth genomic and functional analysis of 1868 high-quality genomes, combining data from public databases with our new isolates. This data set represented 387 colonization-selective species-level clusters, of which eight genera represented multilineage clusters. Pan-genome analysis, single-nucleotide polymorphism (SNP) identification, and probiotic functional predictions revealed that species taxonomy, habitats, and geography together shape the functional diversity of Lachnospiraceae. Moreover, analyses of associations with atherosclerotic cardiovascular disease (ACVD) and inflammatory bowel disease (IBD) indicated that several strains of potentially novel Lachnospiraceae species possess the capacity to reduce the abundance of opportunistic pathogens, thereby imparting potential health benefits. Our findings shed light on the untapped potential of novel species enabling knowledge-based selection of strains for the development of next-generation probiotics holding promise for improving human health and disease management.
Collapse
Affiliation(s)
- Xiaoqian Lin
- BGI Research Shenzhen China
- School of Bioscience and Biotechnology South China University of Technology Guangzhou China
| | | | - Zhinan Wu
- BGI Research Shenzhen China
- College of Life Sciences University of Chinese Academy of Sciences Beijing China
| | | | | | | | - Xudong Liu
- BGI Research Shenzhen China
- College of Life Sciences University of Chinese Academy of Sciences Beijing China
| | - Wenxi Li
- BGI Research Shenzhen China
- School of Bioscience and Biotechnology South China University of Technology Guangzhou China
| | | | | | - Xun Xu
- BGI Research Shenzhen China
| | - Jian Wang
- BGI Research Shenzhen China
- James D. Watson Institute of Genome Sciences Hangzhou China
| | - Huanming Yang
- BGI Research Shenzhen China
- James D. Watson Institute of Genome Sciences Hangzhou China
| | - Karsten Kristiansen
- BGI Research Shenzhen China
- Laboratory of Genomics and Molecular Biomedicine University of Copenhagen Copenhagen Denmark
| | - Liang Xiao
- BGI Research Shenzhen China
- College of Life Sciences University of Chinese Academy of Sciences Beijing China
- Shenzhen Engineering Laboratory of Detection and Intervention of human intestinal microbiome, BGI-Shenzhen Shenzhen China
| | - Yuanqiang Zou
- BGI Research Shenzhen China
- Laboratory of Genomics and Molecular Biomedicine University of Copenhagen Copenhagen Denmark
- Shenzhen Engineering Laboratory of Detection and Intervention of human intestinal microbiome, BGI-Shenzhen Shenzhen China
| |
Collapse
|
10
|
Lordan C, Roche AK, Delsing D, Nauta A, Groeneveld A, MacSharry J, Cotter PD, van Sinderen D. Linking human milk oligosaccharide metabolism and early life gut microbiota: bifidobacteria and beyond. Microbiol Mol Biol Rev 2024; 88:e0009423. [PMID: 38206006 PMCID: PMC10966949 DOI: 10.1128/mmbr.00094-23] [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: 01/12/2024] Open
Abstract
SUMMARYHuman milk oligosaccharides (HMOs) are complex, multi-functional glycans present in human breast milk. They represent an intricate mix of heterogeneous structures which reach the infant intestine in an intact form as they resist gastrointestinal digestion. Therefore, they confer a multitude of benefits, directly and/or indirectly, to the developing neonate. Certain bifidobacterial species, being among the earliest gut colonizers of breast-fed infants, have an adapted functional capacity to metabolize various HMO structures. This ability is typically observed in infant-associated bifidobacteria, as opposed to bifidobacteria associated with a mature microbiota. In recent years, information has been gleaned regarding how these infant-associated bifidobacteria as well as certain other taxa are able to assimilate HMOs, including the mechanistic strategies enabling their acquisition and consumption. Additionally, complex metabolic interactions occur between microbes facilitated by HMOs, including the utilization of breakdown products released from HMO degradation. Interest in HMO-mediated changes in microbial composition and function has been the focal point of numerous studies, in recent times fueled by the availability of individual biosynthetic HMOs, some of which are now commonly included in infant formula. In this review, we outline the main HMO assimilatory and catabolic strategies employed by infant-associated bifidobacteria, discuss other taxa that exhibit breast milk glycan degradation capacity, and cover HMO-supported cross-feeding interactions and related metabolites that have been described thus far.
Collapse
Affiliation(s)
- Cathy Lordan
- Teagasc Food Research Centre, Fermoy, Co Cork, Ireland
| | - Aoife K. Roche
- APC Microbiome Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | | | - Arjen Nauta
- FrieslandCampina, Amersfoort, the Netherlands
| | | | - John MacSharry
- APC Microbiome Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Paul D. Cotter
- Teagasc Food Research Centre, Fermoy, Co Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Douwe van Sinderen
- APC Microbiome Ireland, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
11
|
DuPont HL, Salge MMH. The Importance of a Healthy Microbiome in Pregnancy and Infancy and Microbiota Treatment to Reverse Dysbiosis for Improved Health. Antibiotics (Basel) 2023; 12:1617. [PMID: 37998819 PMCID: PMC10668833 DOI: 10.3390/antibiotics12111617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/05/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND The microbiome of newborn infants during the first 1000 days, influenced early on by their mothers' microbiome health, mode of delivery and breast feeding, orchestrates the education and programming of the infant's immune system and determines in large part the general health of the infant for years. METHODS PubMed was reviewed for maternal infant microbiome health and microbiota therapy in this setting with prebiotics, probiotics, vaginal seeding and fecal microbiota transplantation (FMT). RESULTS A healthy nonobese mother, vaginal delivery and strict breast feeding contribute to microbiome health in a newborn and young infant. With reduced microbiome diversity (dysbiosis) during pregnancy, cesarean delivery, prematurity, and formula feeding contribute to dysbiosis in the newborn. Microbiota therapy is an important approach to repair dysbiosis in pregnant women and their infants. Currently available probiotics can have favorable metabolic effects on mothers and infants, but these effects are variable. In research settings, reversal of infant dysbiosis can be achieved via vaginal seeding or FMT. Next generation probiotics in development should replace current probiotics and FMT. CONCLUSIONS The most critical phase of human microbiome development is in the first 2-3 years of life. Preventing and treating dysbiosis during pregnancy and early life can have a profound effect on an infant's later health.
Collapse
Affiliation(s)
- Herbert L. DuPont
- Division of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas, Houston, TX 77030, USA
- Department of Internal Medicine, University of Texas McGovern Medical School, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Kelsey Research Foundation, Houston, TX 77005, USA
| | | |
Collapse
|
12
|
Krishnamurthy HK, Pereira M, Bosco J, George J, Jayaraman V, Krishna K, Wang T, Bei K, Rajasekaran JJ. Gut commensals and their metabolites in health and disease. Front Microbiol 2023; 14:1244293. [PMID: 38029089 PMCID: PMC10666787 DOI: 10.3389/fmicb.2023.1244293] [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: 06/22/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose of review This review comprehensively discusses the role of the gut microbiome and its metabolites in health and disease and sheds light on the importance of a holistic approach in assessing the gut. Recent findings The gut microbiome consisting of the bacteriome, mycobiome, archaeome, and virome has a profound effect on human health. Gut dysbiosis which is characterized by perturbations in the microbial population not only results in gastrointestinal (GI) symptoms or conditions but can also give rise to extra-GI manifestations. Gut microorganisms also produce metabolites (short-chain fatty acids, trimethylamine, hydrogen sulfide, methane, and so on) that are important for several interkingdom microbial interactions and functions. They also participate in various host metabolic processes. An alteration in the microbial species can affect their respective metabolite concentrations which can have serious health implications. Effective assessment of the gut microbiome and its metabolites is crucial as it can provide insights into one's overall health. Summary Emerging evidence highlights the role of the gut microbiome and its metabolites in health and disease. As it is implicated in GI as well as extra-GI symptoms, the gut microbiome plays a crucial role in the overall well-being of the host. Effective assessment of the gut microbiome may provide insights into one's health status leading to more holistic care.
Collapse
Affiliation(s)
| | | | - Jophi Bosco
- Vibrant America LLC., San Carlos, CA, United States
| | | | | | | | - Tianhao Wang
- Vibrant Sciences LLC., San Carlos, CA, United States
| | - Kang Bei
- Vibrant Sciences LLC., San Carlos, CA, United States
| | | |
Collapse
|
13
|
Huang H, Cheng S, Yang X, Liu L, Cheng B, Meng P, Pan C, Wen Y, Jia Y, Liu H, Zhang F. Dissecting the Association between Gut Microbiota and Brain Structure Change Rate: A Two-Sample Bidirectional Mendelian Randomization Study. Nutrients 2023; 15:4227. [PMID: 37836511 PMCID: PMC10574136 DOI: 10.3390/nu15194227] [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: 08/30/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
The connection between the gut microbiota and brain structure changes is still unclear. We conducted a Mendelian randomization (MR) study to examine the bidirectional causality between the gut microbiota (211 taxa, including 131 genera, 35 families, 20 orders, 16 classes and 9 phyla; N = 18,340 individuals) and age-independent/dependent longitudinal changes in brain structure across the lifespan (N = 15,640 individuals aged 4~99 years). We identified causal associations between the gut microbiota and age-independent/dependent longitudinal changes in brain structure, such as family Peptostreptococcaceae with age-independent longitudinal changes of cortical gray matter (GM) volume and genus Faecalibacterium with age-independent average cortical thickness and cortical GM volume. Taking age-independent longitudinal changes in brain structure across the lifespan as exposures, there were causal relationships between the surface area and genus Lachnospiraceae. Our findings may serve as fundamentals for further research on the genetic mechanisms and biological treatment of complex traits and diseases associated with the gut microbiota and the brain structure change rate.
Collapse
Affiliation(s)
- Huimei Huang
- Department of Nephrology, Xi’an Children’s Hospital, The Affiliated Children’s Hospital of Xi’an Jiaotong University, Xi’an 710003, China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (S.C.); (X.Y.); (L.L.); (B.C.); (P.M.); (C.P.); (Y.W.); (Y.J.); (H.L.)
| | - Xuena Yang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (S.C.); (X.Y.); (L.L.); (B.C.); (P.M.); (C.P.); (Y.W.); (Y.J.); (H.L.)
| | - Li Liu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (S.C.); (X.Y.); (L.L.); (B.C.); (P.M.); (C.P.); (Y.W.); (Y.J.); (H.L.)
| | - Bolun Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (S.C.); (X.Y.); (L.L.); (B.C.); (P.M.); (C.P.); (Y.W.); (Y.J.); (H.L.)
| | - Peilin Meng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (S.C.); (X.Y.); (L.L.); (B.C.); (P.M.); (C.P.); (Y.W.); (Y.J.); (H.L.)
| | - Chuyu Pan
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (S.C.); (X.Y.); (L.L.); (B.C.); (P.M.); (C.P.); (Y.W.); (Y.J.); (H.L.)
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (S.C.); (X.Y.); (L.L.); (B.C.); (P.M.); (C.P.); (Y.W.); (Y.J.); (H.L.)
| | - Yumeng Jia
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (S.C.); (X.Y.); (L.L.); (B.C.); (P.M.); (C.P.); (Y.W.); (Y.J.); (H.L.)
| | - Huan Liu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (S.C.); (X.Y.); (L.L.); (B.C.); (P.M.); (C.P.); (Y.W.); (Y.J.); (H.L.)
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, China; (S.C.); (X.Y.); (L.L.); (B.C.); (P.M.); (C.P.); (Y.W.); (Y.J.); (H.L.)
| |
Collapse
|
14
|
Abdugheni R, Liu C, Liu FL, Zhou N, Jiang CY, Liu Y, Li L, Li WJ, Liu SJ. Comparative genomics reveals extensive intra-species genetic divergence of the prevalent gut commensal Ruminococcus gnavus. Microb Genom 2023; 9:mgen001071. [PMID: 37486746 PMCID: PMC10438805 DOI: 10.1099/mgen.0.001071] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Ruminococcus gnavus is prevalent in the intestines of humans and animals, and ambiguities have been reported regarding its relations with the development of diseases and host well-being. We postulate the ambiguities of its function in different cases may be attributed to strain-level variability of genomic features of R. gnavus. We performed comparative genomic and pathogenicity prediction analysis on 152 filtered high-quality genomes, including 4 genomes of strains isolated from healthy adults in this study. The mean G+C content of genomes of R. gnavus was 42.73±0.33 mol%, and the mean genome size was 3.46±0.34 Mbp. Genome-wide evolutionary analysis revealed R. gnavus genomes were divided into three major phylogenetic clusters. Pan-core genome analysis revealed that there was a total of 28 072 predicted genes, and the core genes, soft-core genes, shell genes and cloud genes accounted for 3.74 % (1051/28 072), 1.75 % (491/28 072), 9.88 % (2774/28 072) and 84.63 % (23 756/28 072) of the total genes, respectively. The small proportion of core genes reflected the wide divergence among R. gnavus strains. We found certain coding sequences with determined health benefits (such as vitamin production and arsenic detoxification), whilst some had an implication of health adversity (such as sulfide dehydrogenase subunits). The functions of the majority of core genes were unknown. The most widespread genes functioning in antibiotic resistance and virulence are tetO (tetracycline-resistance gene, present in 75 strains) and cps4J (capsular polysaccharide biosynthesis protein Cps4J encoding gene, detected in 3 genomes), respectively. Our results revealed genomic divergence and the existence of certain safety-relevant factors of R. gnavus. This study provides new insights for understanding the genomic features and health relevance of R. gnavus, and raises concerns regarding predicted prevalent pathogenicity and antibiotic resistance among most of the strains.
Collapse
Affiliation(s)
- Rashidin Abdugheni
- State Key Laboratory of Desert and Oasis Ecology, Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, PR China
| | - Chang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266000, PR China
| | - Feng-Lan Liu
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
- College of Life Sciences, Hebei University, Baoding 071000, PR China
| | - Nan Zhou
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Cheng-Ying Jiang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266000, PR China
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
- University of the Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yonghong Liu
- State Key Laboratory of Desert and Oasis Ecology, Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, PR China
| | - Li Li
- State Key Laboratory of Desert and Oasis Ecology, Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, PR China
| | - Wen-Jun Li
- State Key Laboratory of Desert and Oasis Ecology, Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, PR China
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resources and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, PR China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266000, PR China
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, PR China
- University of the Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
15
|
Li X, Chen Y, Song L, Wang J, Song Z, Zhao X, Zhou C, Wu Y. Partial enzymolysis affects the digestion of tamarind seed polysaccharides in vitro: Degradation accelerates and gut microbiota regulates. Int J Biol Macromol 2023; 237:124175. [PMID: 37003195 DOI: 10.1016/j.ijbiomac.2023.124175] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023]
Abstract
Two hydrolyzed fractions of tamarind seed polysaccharide (TSP), denoted ETSP1 (176.68 kDa) and ETSP2 (34.34 kDa), were prepared by partial degradation via endo-xyloglucanase, and then characterized and evaluated by simulated gastrointestinal digestion in vitro. The results showed that the hydrolyzed TSPs remained indigestible in gastric and small intestinal media, and were fermented by gut microbiota, similar to the native TSP (Mw = 481.52 kDa). Although the degradation of hydrolyzed TSPs was accelerated during fermentation with a decreasing degree of polymerization, the content of produced total short-chain fatty acids (SCFAs) decreased. After fermentation, the gut microbiota composition was modified, esp. the Firmicutes/Bacteroidetes ratio decreased (1.06 vs. 0.96 vs. 0.80) with a decreasing degree of polymerization, which implied that the potential anti-obesity prebiotic effect was enhanced. At the genus level, hydrolyzed TSPs maintained similar roles as native TSP, including promoting beneficial bacteria (Bifidobacterium, Parabacteroides, and Faecalibacterium) and inhibiting enteropathogenic bacteria (Escherichia-Shigella and Dorea). Moreover, ETSP1 had additional potential due to abundant Bacteroides vulgatus (LDA = 4.68), and ETSP2 might perform better as related to Bacteroides xylanisolvens (LDA = 4.40). All these results indicated the prebiotic potential of hydrolyzed TSP with detailed information about changes in degradation and gut microbiota based on enzyme-hydrolysis.
Collapse
Affiliation(s)
- Xujiao Li
- Institute for Agro-food Standards and Testing Technology, Laboratory of Quality & Safety Risk Assessment for Agro-products (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China; Shanghai Engineering Research Center of Food Safety, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yinan Chen
- Shanghai Engineering Research Center of Food Safety, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Lihua Song
- Shanghai Engineering Research Center of Food Safety, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Jiangmei Wang
- Weifang Ecological Environment Monitoring Center, Weifang 261041, China.
| | - Zibo Song
- Yunnan Maoduoli Group Food Co., Ltd., Yuxi 653100, China.
| | - Xiaoyan Zhao
- Institute for Agro-food Standards and Testing Technology, Laboratory of Quality & Safety Risk Assessment for Agro-products (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China.
| | - Changyan Zhou
- Institute for Agro-food Standards and Testing Technology, Laboratory of Quality & Safety Risk Assessment for Agro-products (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China.
| | - Yan Wu
- Shanghai Engineering Research Center of Food Safety, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
16
|
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
|
17
|
Lee S, Choi A, Park KH, Lee S, Yoon H, Kim P. Single-cell hemoprotein (heme-SCP) exerts the prebiotic potential to establish a healthy gut microbiota in small pet dogs. Food Sci Biotechnol 2023; 32:489-496. [PMID: 36911324 PMCID: PMC9992493 DOI: 10.1007/s10068-022-01195-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/08/2022] [Accepted: 10/24/2022] [Indexed: 11/25/2022] Open
Abstract
To investigate the effect of the single-cell hemoprotein (heme-SCP) source on animals, a dog-treat (100 g for each dog) harboring 0.2% heme-SCP was manufactured and fed to seven pet dogs (< 10 kg) in a randomized manner (irrespective of owner's feeding style, dogs' health conditions, and staple diets), and the feces before and after the dog-treat diet were analyzed to define the structure of the microbiota. The total bacterial species of the seven dogs showed no difference (564-584), although the bacterial compositions varied significantly. The Firmicutes phylum increased (54.7-73.7%), showing differential species composition before and after heme-SCP intake. Proteobacteria, Bacteroidetes, and Fusobacteria decreased (5.4-3.8%, 32.9-16.8%, and 6.3-3.6%, respectively), which agreed with the previous observation of deliberate feeding. Therefore, it is conceivable that heme-SCP as a prebiotic can shape the gut microbiota regardless of the administration method. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-022-01195-9.
Collapse
Affiliation(s)
- Seungki Lee
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon, Gyeonggi 14662 South Korea
| | - Ahyoung Choi
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon, Gyeonggi 14662 South Korea
| | | | - Seoyeon Lee
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499 South Korea
| | - Hyunjin Yoon
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Yeongtong-gu, Suwon, Gyeonggi 16499 South Korea
| | - Pil Kim
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Wonmi-gu, Bucheon, Gyeonggi 14662 South Korea.,HemoLab Ltd. Co., Bucheon, 14622 South Korea
| |
Collapse
|
18
|
Early life gut microbiota profiles linked to synbiotic formula effects: a randomized clinical trial in European infants. Am J Clin Nutr 2023; 117:326-339. [PMID: 36811568 DOI: 10.1016/j.ajcnut.2022.11.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 11/05/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Microbial colonization of the gastrointestinal tract after birth is an essential event that influences infant health with life-long consequences. Therefore, it is important to investigate strategies to positively modulate colonization in early life. OBJECTIVES This randomized, controlled intervention study included 540 infants to investigate the effects of a synbiotic intervention formula (IF) containing Limosilactobacillus fermentum CECT5716 and galacto-oligosaccharides on the fecal microbiome. METHODS The fecal microbiota from infants was analyzed by 16S rRNA amplicon sequencing at 4, 12, and 24 months of age. Metabolites (e.g., short-chain fatty acids) and other milieu parameters (e.g., pH, humidity, and IgA) were also measured in stool samples. RESULTS Microbiota profiles changed with age, with major differences in diversity and composition. Significant effects of the synbiotic IF compared with control formula (CF) were visible at month 4, including higher occurrence of Bifidobacterium spp. and Lactobacillaceae and lower occurrence of Blautia spp., as well as Ruminoccocus gnavus and relatives. This was accompanied by lower fecal pH and concentrations of butyrate. After de novo clustering at 4 months of age, overall phylogenetic profiles of the infants receiving IF were closer to reference profiles of those fed with human milk than infants fed CF. The changes owing to IF were associated with fecal microbiota states characterized by lower occurrence of Bacteroides compared with higher levels of Firmicutes (valid name Bacillota), Proteobacteria (valid name Pseudomonadota), and Bifidobacterium at 4 months of age. These microbiota states were linked to higher prevalence of infants born by Cesarean section. CONCLUSIONS The synbiotic intervention influenced fecal microbiota and milieu parameters at an early age depending on the overall microbiota profiles of the infants, sharing a few similarities with breastfed infants. This trial was registered at clinicaltrials.gov as NCT02221687.
Collapse
|
19
|
Roager HM, Stanton C, Hall LJ. Microbial metabolites as modulators of the infant gut microbiome and host-microbial interactions in early life. Gut Microbes 2023; 15:2192151. [PMID: 36942883 PMCID: PMC10038037 DOI: 10.1080/19490976.2023.2192151] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023] Open
Abstract
The development of infant gut microbiome is a pivotal process affecting the ecology and function of the microbiome, as well as host health. While the establishment of the infant microbiome has been of interest for decades, the focus on gut microbial metabolism and the resulting small molecules (metabolites) has been rather limited. However, technological and computational advances are now enabling researchers to profile the plethora of metabolites in the infant gut, allowing for improved understanding of how gut microbial-derived metabolites drive microbiome community structuring and host-microbial interactions. Here, we review the current knowledge on development of the infant gut microbiota and metabolism within the first year of life, and discuss how these microbial metabolites are key for enhancing our basic understanding of interactions during the early life developmental window.
Collapse
Affiliation(s)
- Henrik M. Roager
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg, Denmark
| | - Catherine Stanton
- APC Microbiome Ireland, Teagasc Moorepark Food Research Centre, Fermoy, Co. Cork, Ireland
| | - Lindsay J. Hall
- Gut Microbes & Health, Quadram Institute Biosciences, Norwich, UK
- Intestinal Microbiome, School of Life Sciences, ZIEL – Institute for Food & Health, Technical University of Munich, Freising, Germany
- Norwich Medical School, University of East Anglia, Norwich, UK
| |
Collapse
|
20
|
Abdugheni R, Wang W, Wang Y, Du M, Liu F, Zhou N, Jiang C, Wang C, Wu L, Ma J, Liu C, Liu S. Metabolite profiling of human-originated Lachnospiraceae at the strain level. IMETA 2022; 1:e58. [PMID: 38867908 PMCID: PMC10989990 DOI: 10.1002/imt2.58] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 06/14/2024]
Abstract
The human gastrointestinal (GI) tract harbors diverse microbes, and the family Lachnospiraceae is one of the most abundant and widely occurring bacterial groups in the human GI tract. Beneficial and adverse effects of the Lachnospiraceae on host health were reported, but the diversities at species/strain levels as well as their metabolites of Lachnospiraceae have been, so far, not well documented. In the present study, we report on the collection of 77 human-originated Lachnospiraceae species (please refer hLchsp, https://hgmb.nmdc.cn/subject/lachnospiraceae) and the in vitro metabolite profiles of 110 Lachnospiraceae strains (https://hgmb.nmdc.cn/subject/lachnospiraceae/metabolites). The Lachnospiraceae strains in hLchsp produced 242 metabolites of 17 categories. The larger categories were alcohols (89), ketones (35), pyrazines (29), short (C2-C5), and long (C > 5) chain acids (31), phenols (14), aldehydes (14), and other 30 compounds. Among them, 22 metabolites were aromatic compounds. The well-known beneficial gut microbial metabolite, butyric acid, was generally produced by many Lachnospiraceae strains, and Agathobacter rectalis strain Lach-101 and Coprococcus comes strain NSJ-173 were the top 2 butyric acid producers, as 331.5 and 310.9 mg/L of butyric acids were produced in vitro, respectively. Further analysis of the publicly available cohort-based volatile-metabolomic data sets of human feces revealed that over 30% of the prevailing volatile metabolites were covered by Lachnospiraceae metabolites identified in this study. This study provides Lachnospiraceae strain resources together with their metabolic profiles for future studies on host-microbe interactions and developments of novel probiotics or biotherapies.
Collapse
Affiliation(s)
- Rashidin Abdugheni
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Desert and Oasis EcologyXinjiang Institute of Ecology and Geography, Chinese Academy of SciencesUrumqiChina
| | - Wen‐Zhao Wang
- State Key Laboratory of MycologyInstitute of Microbiology, Chinese Academy of SciencesBeijingChina
| | - Yu‐Jing Wang
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Meng‐Xuan Du
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| | - Feng‐Lan Liu
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- College of Life SciencesHebei UniversityBaodingChina
| | - Nan Zhou
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
| | - Cheng‐Ying Jiang
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Chang‐Yu Wang
- Colleg of Life SciencesUniversity of Science and Technology of ChinaHefeiChina
| | - Linhuan Wu
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
| | - Juncai Ma
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
| | - Chang Liu
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| | - Shuang‐Jiang Liu
- State Key Laboratory of Microbial Resources, Environmental Microbiology Research Center (EMRC)Institute of Microbiology, Chinese Academy of SciencesBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| |
Collapse
|
21
|
A Multiscale Spatiotemporal Model Including a Switch from Aerobic to Anaerobic Metabolism Reproduces Succession in the Early Infant Gut Microbiota. mSystems 2022; 7:e0044622. [PMID: 36047700 PMCID: PMC9600552 DOI: 10.1128/msystems.00446-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The human intestinal microbiota starts to form immediately after birth and is important for the health of the host. During the first days, facultatively anaerobic bacterial species generally dominate, such as Enterobacteriaceae. These are succeeded by strictly anaerobic species, particularly Bifidobacterium species. An early transition to Bifidobacterium species is associated with health benefits; for example, Bifidobacterium species repress growth of pathogenic competitors and modulate the immune response. Succession to Bifidobacterium is thought to be due to consumption of intracolonic oxygen present in newborns by facultative anaerobes, including Enterobacteriaceae. To study if oxygen depletion suffices for the transition to Bifidobacterium species, here we introduced a multiscale mathematical model that considers metabolism, spatial bacterial population dynamics, and cross-feeding. Using publicly available metabolic network data from the AGORA collection, the model simulates ab initio the competition of strictly and facultatively anaerobic species in a gut-like environment under the influence of lactose and oxygen. The model predicts that individual differences in intracolonic oxygen in newborn infants can explain the observed individual variation in succession to anaerobic species, in particular Bifidobacterium species. Bifidobacterium species became dominant in the model by their use of the bifid shunt, which allows Bifidobacterium to switch to suboptimal yield metabolism with fast growth at high lactose concentrations, as predicted here using flux balance analysis. The computational model thus allows us to test the internal plausibility of hypotheses for bacterial colonization and succession in the infant colon. IMPORTANCE The composition of the infant microbiota has a great impact on infant health, but its controlling factors are still incompletely understood. The frequently dominant anaerobic Bifidobacterium species benefit health, e.g., they can keep harmful competitors under control and modulate the intestinal immune response. Controlling factors could include nutritional composition and intestinal mucus composition, as well as environmental factors, such as antibiotics. We introduce a modeling framework of a metabolically realistic intestinal microbial ecology in which hypothetical scenarios can be tested and compared. We present simulations that suggest that greater levels of intraintestinal oxygenation more strongly delay the dominance of Bifidobacterium species, explaining the observed variety of microbial composition and demonstrating the use of the model for hypothesis generation. The framework allowed us to test a variety of controlling factors, including intestinal mixing and transit time. Future versions will also include detailed modeling of oligosaccharide and mucin metabolism.
Collapse
|
22
|
Gut microbiota: a new avenue to reveal pathological mechanisms of constipation. Appl Microbiol Biotechnol 2022; 106:6899-6913. [PMID: 36190540 DOI: 10.1007/s00253-022-12197-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/27/2022]
Abstract
Constipation is very pervasive all over the world. It is a common multifactorial gastrointestinal disease, and its etiology and pathomechanism are not completely clear. Now, increasing evidence shows that intestinal flora is closely related to constipation. Intestinal flora is the largest microbiota in the human body and has powerful metabolic functions. Intestinal flora can produce a variety of metabolites, such as bile acids, short-chain fatty acids, tryptophan metabolites, and methane, which have important effects on intestinal motility and secretion. The host can also monitor the intestinal flora and regulate gut dysbacteriosis in constipation. To explore the relationship between intestinal flora and host, the combination of multiomics technology has become the powerful and effective method. Furthermore, the homeostasis restoration of intestinal flora also provides a new strategy for the treatment of constipation. This review aims to explore the interaction between intestinal flora and host in constipation, which contributes to disclose the pathogenesis of constipation and the development of novel drugs for the treatment of constipation from the perspective of intestinal flora. KEY POINTS: • This review highlights the regulation of gut microbiota on the intestinal motility and secretion of host. • The current review gives an insight into the role of the host on the recognition and regulation of intestinal ecology under constipation. • The article also introduces some novel methods of current gut microbiota research and gut microbiota-based constipation therapies.
Collapse
|
23
|
Liu X, Li J, Hao L, Degen A, Wang D, Ma Y, Niu J, Cheng Y, Liu S. Effect of the ratio of dietary metabolizable energy to nitrogen content on production performance, serum metabolites, rumen fermentation parameters, and bacterial diversity in yaks. Front Microbiol 2022; 13:1013980. [PMID: 36304954 PMCID: PMC9593094 DOI: 10.3389/fmicb.2022.1013980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
This study examined the effect of the ratio of dietary metabolizable energy (MJ) to nitrogen (g) content (ME:N) on average daily gain (ADG), blood biochemical indices, rumen fermentation parameters, and rumen bacterial community in yaks. Thirty-six male yaks, aged 2–3 years, were divided into three groups and received a ME:N ratio of 0.42 (HY), 0.36 (MY,) or 0.32 (LY) MJ/g. Dry matter intake ranged between 3.16 and 3.63 kg/d and was lesser (p < 0.001) in the LY group than the other two groups. ME intake increased (p < 0.001) with an increase in the ME:N ratio, while N intake did not differ among groups. The ADG was 660 g/day for the MY group, which was higher (p < 0.005) than the 430 g/day in the LY group, while the HY group gained 560 g/day and did not differ from the other two groups. Feed intake to ADG ratio ranged between 5.95 and 7.95, and numerically was highest in the LY group and lowest in the MY group. In general, the concentration of ruminal total volatile fatty acids (p < 0.03) and molar proportions of propionate (p < 0.04), increased, while the molar proportion of acetate (p < 0.005) and the acetate:propionate ratio decreased (p < 0.001) with a decrease in the ME:N ratio. The molar proportion of butyrate did not differ among groups (p = 0.112). Group MY had higher ruminal NH3-N content than group HY and had a higher serum glucose content but lower urea content, lactate dehydrogenase, and creatine kinase content than group LY. In ruminal bacteria at the phylum level, the relative abundance of Firmicutes (F) was greater and of Bacteroidetes (B) was lesser, while the F:B ratio was greater in group MY than in groups HY an LY. We concluded that the yaks consuming the diet containing a ME:N ratio of 0.36 MJ/g had the best performance of the three groups.
Collapse
Affiliation(s)
- Xiaojing Liu
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Yanfen Cheng,
| | - Jie Li
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Science and Veterinary Medicine of Qinghai University, Xining, China
- Gansu Polytechnic College of Animal Husbandry & Engineering, Wuwei, China
- *Correspondence: Yanfen Cheng,
| | - Lizhuang Hao
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Science and Veterinary Medicine of Qinghai University, Xining, China
- Lizhuang Hao,
| | - Allan Degen
- Desert Animal Adaptations and Husbandry, Wyler Department of Dryland Agriculture, Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Dongyang Wang
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Yonggui Ma
- Academy of Plateau Science and Sustainability, People’s Government of Qinghai Province and Beijing Normal University, Key Laboratory of Medicinal Animal and Plant Resources of Qinghai-Tibetan Plateau in Qinghai Province, College of Life Science, Qinghai Normal University, Xining, China
| | - Jianzhang Niu
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Science and Veterinary Medicine of Qinghai University, Xining, China
| | - Yanfen Cheng
- Laboratory of Gastrointestinal Microbiology, National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
- State Key Laboratory of Grassland Agro-Ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
- *Correspondence: Yanfen Cheng,
| | - Shujie Liu
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Academy of Animal Science and Veterinary Medicine of Qinghai University, Xining, China
| |
Collapse
|
24
|
Ahmad AA, Zhang J, Liang Z, Du M, Yang Y, Zheng J, Yan P, Long R, Tong B, Han J, Ding X. Age-dependent variations in rumen bacterial community of Mongolian cattle from weaning to adulthood. BMC Microbiol 2022; 22:213. [PMID: 36071396 PMCID: PMC9450343 DOI: 10.1186/s12866-022-02627-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/25/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Rumen microbes play an important role in ruminant energy supply and animal performance. Previous studies showed that the rumen microbiome of Mongolian cattle has adapted to degrade the rough forage to provide sufficient energy to tolerate the harsh desert ecological conditions. However, little is known about the succession of rumen microbes in different developmental stages of post-weaning Mongolian cattle. METHODS Here, we examined the succession of the rumen microbial composition and structure of 15 post-weaning Mongolian cattle at three developmental stages i.e., 5 months (RM05), 18 months (RM18) and, 36 months (RM36) by using the 16S rRNA gene sequencing method. RESULTS We did not find any age-dependent variations in the ruminal concentrations of any volatile fatty acid (VFA) of Mongolian cattle. The diversity of the rumen bacterial community was significantly lower in RM05 group, which reached to stability with age. Bacteroidetes and Firmicutes were the two dominant phyla among all age groups. Phylum Actinobacteria was significantly higher in RM05 group, phyla Spirochaetes, and Tenericutes were highly abundant in RM18 group, and phyla Proteobacteria and Epsilonbacteraeota were enriched in RM36 group. Genera Prevotella_1, Bacteroides, and Bifidobacterium were abundant in RM05 group. The short chain fatty acid (SCFA) producing bacteria Rikenellaceae_RC9_gut_group showed high abundance in RM18 group and fiber degrading genus Alloprevotella was highly abundant in RM36 group. Random forest analysis identified Alloprevotella, Ileibacterium, and Helicobacter as important age discriminatory genera. In particular, the genera Ruminococcaceae_UCG-005, Bacteroides, Saccharofermentans, and Fibrobacter in RM05, genera [Eubacterium] coprostanoligenes_group, Erysipelotrichaceae_UCG-004, Helicobacter, Saccharofermentans, Papillibacter, and Turicibacter in RM18, and genera Rikenellaceae_RC9_gut_group, Lachnospiraceae_AC2044_group, and Papillibacter in RM36 showed the top interactions values in the intra-group interaction network. CONCLUSIONS The results showed that rumen microbiota of Mongolian cattle reached to stability and maturity with age after weaning. This study provides some theoretical evidence about the importance of functional specific rumen bacteria in different age groups. Further studies are needed to determine their actual roles and interactions with the host.
Collapse
Affiliation(s)
- Anum Ali Ahmad
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, 730050, China.,State Key Laboratory of Grassland Agro-Ecosystems, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jianbo Zhang
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, 730050, China.,Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, 730050, China
| | - Zeyi Liang
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, 730050, China
| | - Mei Du
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, 730050, China
| | - Yayuan Yang
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, 730050, China
| | - Juanshan Zheng
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, 730050, China
| | - Ping Yan
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, 730050, China
| | - RuiJun Long
- State Key Laboratory of Grassland Agro-Ecosystems, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Bin Tong
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, The Research Center for Laboratory Animal Science, School of Life Sciences, Inner Mongolia University, Mongolia, China
| | - Jianlin Han
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China. .,Livestock Genetics Program, International Livestock Research Institute (ILRI), Nairobi, Kenya.
| | - Xuezhi Ding
- Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, 730050, China. .,Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agricultural and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Science, Lanzhou, 730050, China.
| |
Collapse
|
25
|
Lin H, Eggesbø M, Peddada SD. Linear and nonlinear correlation estimators unveil undescribed taxa interactions in microbiome data. Nat Commun 2022; 13:4946. [PMID: 35999204 PMCID: PMC9399263 DOI: 10.1038/s41467-022-32243-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/20/2022] [Indexed: 12/04/2022] Open
Abstract
It is well-known that human gut microbiota form an ecosystem where microbes interact with each other. Due to complex underlying interactions, some microbes may correlate nonlinearly. There are no measures in the microbiome literature we know of that quantify these nonlinear relationships. Here, we develop a methodology called Sparse Estimation of Correlations among Microbiomes (SECOM) for estimating linear and nonlinear relationships among microbes while maintaining the sparsity. SECOM accounts for both sample and taxon-specific biases in its model. Its statistical properties are evaluated analytically and by comprehensive simulation studies. We test SECOM in two real data sets, namely, forehead and palm microbiome data from college-age adults, and Norwegian infant gut microbiome data. Given that forehead and palm are related to skin, as desired, SECOM discovers each genus to be highly correlated between the two sites, but that is not the case with any of the competing methods. It is well-known that infant gut evolves as the child grows. Using SECOM, for the first time in the literature, we characterize temporal changes in correlations among bacterial families during a baby's first year after birth.
Collapse
Affiliation(s)
- Huang Lin
- Biostatistics and Bioinformatics Branch, Eunice Shriver Kennedy NICHD, NIH, Bethesda, MD, USA
| | | | - Shyamal Das Peddada
- Biostatistics and Bioinformatics Branch, Eunice Shriver Kennedy NICHD, NIH, Bethesda, MD, USA.
| |
Collapse
|
26
|
Silverman GJ, Deng J, Azzouz DF. Sex-dependent Lupus Blautia (Ruminococcus) gnavus strain induction of zonulin-mediated intestinal permeability and autoimmunity. Front Immunol 2022; 13:897971. [PMID: 36032126 PMCID: PMC9405438 DOI: 10.3389/fimmu.2022.897971] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Imbalances in the gut microbiome are suspected contributors to the pathogenesis of Systemic Lupus Erythematosus, and our studies and others have documented that patients with active Lupus nephritis have expansions of the obligate anaerobe, Blautia (Ruminococcus) gnavus (RG). To investigate whether the RG strains in Lupus patients have in vivo pathogenic properties in a gnotobiotic system, we colonized C57BL/6 mice with individual RG strains from healthy adults or those from Lupus patients. These strains were similar in their capacity for murine intestinal colonization of antibiotic-preconditioned specific-pathogen-free, as well as of germ-free adults and of their neonatally colonized litters. Lupus-derived RG strains induced high levels of intestinal permeability that was significantly greater in female than male mice, whereas the RG species-type strain (ATCC29149/VPI C7-1) from a healthy donor had little or no effects. These Lupus RG strain-induced functional alterations were associated with RG translocation to mesenteric lymph nodes, and raised serum levels of zonulin, a regulator of tight junction formation between cells that form the gut barrier. Notably, the level of Lupus RG-induced intestinal permeability was significantly correlated with serum IgG anti RG cell-wall lipoglycan antibodies, and with anti-native DNA autoantibodies that are a biomarker for SLE. Strikingly, gut permeability was completely reversed by oral treatment with larazotide acetate, an octapeptide that is a specific molecular antagonist of zonulin. Taken together, these studies document a pathway by which RG strains from Lupus patients contribute to a leaky gut and features of autoimmunity implicated in the pathogenesis of flares of clinical Lupus disease.
Collapse
|
27
|
Wang T, Guan K, Su Q, Wang X, Yan Z, Kuang K, Wang Y, Zhang Q, Zhou X, Liu B. Change of Gut Microbiota in PRRSV-Resistant Pigs and PRRSV-Susceptible Pigs from Tongcheng Pigs and Large White Pigs Crossed Population upon PRRSV Infection. Animals (Basel) 2022; 12:ani12121504. [PMID: 35739841 PMCID: PMC9219425 DOI: 10.3390/ani12121504] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/28/2022] [Accepted: 06/02/2022] [Indexed: 12/05/2022] Open
Abstract
Simple Summary The gut microbiota could directly induce immune responses and affect the health of the host. In this study, we assessed changes in the gut microbiota of resistant segregated phenotypic pigs under Porcine Reproductive and Respiratory Syndrome Virus exposure. The results showed that the resistance of pigs was related to the composition of gut microbiota. The quantity and relative abundance of probiotics in resistant individuals positively affected host immunity and growth performance, whereas high levels of pathogenic bacteria in susceptible individuals were associated with poorer clinical outcomes. The results of this study suggest that gut microbiota may serve as an effective probiotic resource to provide new methods for PRRS prevention and treatment. Abstract Porcine Reproductive and Respiratory Syndrome (PRRS) is one of the serious infectious diseases that threatens the swine industry. Increasing evidence shows that gut microbiota plays an important role in regulating host immune responses to PRRS virus (PRRSV). The aim of this study was to investigate gut microbiota difference between PRRSV-resistant pigs and PRRSV-suspectable pigs derived from a Tongcheng pigs and Large White pigs crossed population. PRRSV infection induces an increase in the abundance and diversity of gut microbiota. Correlation analysis showed that 36 genera were correlated with viral loads or weight gain after PRRSV infection. Prevotellaceae-NK3B31-group, Christensenellaceae-R7-group, and Parabacteroides were highly correlated with both viral load and weight gain. Notably, the diversity and abundance of beneficial bacteria such as Prevotellaceae-NK3B31-group was high in resistant pigs, and the diversity and abundance of pathogenic bacteria such as Campylobacter and Desulfovibrio were high in susceptible pigs. Gut microbiota were significantly associated with immune function and growth performance, suggesting that these genera might be related to viremia, clinical symptoms, and disease resistance. Altogether, this study revealed the correlation of gut microbiota with PRRSV infection and gut microbiota interventions may provide an effective prevention against PRRSV infection.
Collapse
Affiliation(s)
- Tengfei Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (T.W.); (K.G.); (Q.S.); (X.W.); (Z.Y.); (K.K.); (Y.W.)
| | - Kaifeng Guan
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (T.W.); (K.G.); (Q.S.); (X.W.); (Z.Y.); (K.K.); (Y.W.)
| | - Qiuju Su
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (T.W.); (K.G.); (Q.S.); (X.W.); (Z.Y.); (K.K.); (Y.W.)
| | - Xiaotong Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (T.W.); (K.G.); (Q.S.); (X.W.); (Z.Y.); (K.K.); (Y.W.)
| | - Zengqiang Yan
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (T.W.); (K.G.); (Q.S.); (X.W.); (Z.Y.); (K.K.); (Y.W.)
| | - Kailin Kuang
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (T.W.); (K.G.); (Q.S.); (X.W.); (Z.Y.); (K.K.); (Y.W.)
| | - Yuan Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (T.W.); (K.G.); (Q.S.); (X.W.); (Z.Y.); (K.K.); (Y.W.)
| | - Qingde Zhang
- Laboratory Animal Centre, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Xiang Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (T.W.); (K.G.); (Q.S.); (X.W.); (Z.Y.); (K.K.); (Y.W.)
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- The Engineering Technology Research Center of Hubei Province Local Pig Breed Improvement, Wuhan 430070, China
- Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence: (X.Z.); (B.L.)
| | - Bang Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (T.W.); (K.G.); (Q.S.); (X.W.); (Z.Y.); (K.K.); (Y.W.)
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
- The Engineering Technology Research Center of Hubei Province Local Pig Breed Improvement, Wuhan 430070, China
- Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
- Correspondence: (X.Z.); (B.L.)
| |
Collapse
|
28
|
He X, Chen D, Guo Y, Zhang X, Ma Y, Zhao S. Walnut Meal Extracts Rich In Polyphenols Mitigate Insulin Resistance and Modulate Gut Microbiota in High Fat Diet-Fed Rats. J Med Food 2022; 25:618-629. [PMID: 35708635 DOI: 10.1089/jmf.2021.k.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Walnut kernel is a traditional Chinese herb recorded in the Chinese Pharmacopoeia with the efficacies of invigorating kidney, tonifying lung, and relaxing bowel. However, the potential mechanisms were unclear. This article aims to uncover the interdict mechanisms of walnut meal extracts (WMP) on high-fat diet (HFD) induced metabolic disorders in rats and reveal how the WMP benefits are associated with changes in the intestinal flora. Sprague-Dawley (SD) rats were fed a standard chow diet or an HFD for 18 weeks. After 6 weeks, the HFD rats were supplemented with 750 mg WMP/kg body weight or the vehicle for 12 weeks. The structure of gut microbiota was assessed by analyzing 16S rDNA sequences. WMP suppressed the weight gain and visceral obesity. WMP treatment also improved lipid profiles and increased antioxidative activities. WMP fully reversed hepatic steatosis with the upregulation of adipocytokines involved in lipid catabolism (e.g., adiponectin, PPAR-γ, visfatin, CEBPα) and the increased activities of lipoprotein lipase and hormone-sensitive lipase, which were associated with glucose tolerance improvement and insulin resistance (IR) mitigation. As revealed by 16S rDNA sequencing, WMP restored the diversity of intestinal flora reduced by HFD. WMP dramatically reduced the abundance of Gram-negative bacteria, especially Fusobacterium varium and Bacteroides vulgatus, and sharply increased the abundance of Lactobacillus animalis decreased by HFD. Our findings demonstrated that WMP suppressed the weight gain and adiposity in HFD-fed rats and fully reversed HFD induced IR and hepatic steatosis while dramatically reducing the abundance of Fusobacteriaceae and Enterobacteriaceae, underscoring the gut-liver axis as a primary target of walnut polyphenols.
Collapse
Affiliation(s)
- Xingping He
- Faculty of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, China.,Faculty of Medicine, Lijiang Culture and Tourism College, Lijiang, China
| | - Dan Chen
- Department of Quality Control, Yunnan Institute of Tobacco Quality Inspection and Supervision, Kunming, China
| | - Yan Guo
- Faculty of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Xi Zhang
- Faculty of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Yage Ma
- Faculty of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Shenglan Zhao
- Faculty of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, China
| |
Collapse
|
29
|
Abdugheni R, Wang YJ, Li DH, Du MX, Liu C, Zhou N, Liu SJ. Pararoseburia lenta gen. nov., sp. nov. isolated from human faeces. Int J Syst Evol Microbiol 2022; 72. [DOI: 10.1099/ijsem.0.005371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
A strictly anaerobic, motile bacterium, designated as strain NSJ-9T, was isolated from human faeces. Cells were Gram-negative, non-spore-forming, non-pigmented, and spiral-shaped or slightly curved rods with flagella. Optimal growth in M2GSC medium was observed at 37 °C (growth range 30–45 °C) and pH 6.5–7.0 (growth range 6.5–7.5) under anaerobic conditions. Phylogenetic analysis of the 16S rRNA gene revealed that strain NSJ-9T formed a distinct phylogenetic lineage that reflects a new genus in the family
Lachnospiraceae
, with high levels of similarity to
Roseburia hominis
A2-183T (95.2 %),
Roseburia cecicola
ATCC 33874T (95.2 %),
Pseudobutyrivibrio ruminis
DSM 9787T (95.2 %),
Pseudobutyrivibrio xylanivorans
MZ 5T (94.8%) and
Roseburia faecis
M72/1T (94.4 %). Genomic similarity (average nucleotide identity and digital DNA–DNA hybridization) values between strain NSJ-9T and its phylogenetic neighbours were below 71 and 31 %, respectively, indicating that strain NSJ-9T represented a novel species. The average amino acid identity and the percentage of conserved proteins between strain NSJ-9T and other related members of the family
Lachnospiraceae
were below 63 and 50 %, respectively, supporting that strain NSJ-9T was a member of a new genus. The predominant cellular fatty acids of strain NSJ-9T were C16 : 0 and C17 : 0 2-OH, and major polar lipids were glycolipids. The end products of glucose fermentation were acetate, propionate, iso-butyrate, butyrate and valerate. Phylogenetic and phylogenomic lineage, pairwise determined genome identity analysis suggested that strain NSJ-9T represents a novel genus in the family
Lachnospiraceae
. The genome size of strain NSJ-9T is 2.56 Mbp with 44.9 mol% G+C content. Collectively, the genotypic and phenotypic differences between phylogenetic relatives suggested strain NSJ-9T represented a novel species of a new genus, for which the name Pararoseburia lenta gen. nov., sp. nov. is proposed. The type strain of Pararoseburia lenta is NSJ-9T (=CGMCC 1.32469T=KCTC 15957T).
Collapse
Affiliation(s)
- Rashidin Abdugheni
- University of Chinese Academy of Sciences, Beijing 100049, PR China
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Yu-Jing Wang
- University of Chinese Academy of Sciences, Beijing 100049, PR China
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Dan-Hua Li
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Meng-Xuan Du
- State Key Laboratory of Microbial Biotechnology, Shandong University, Qingdao 266237, PR China
| | - Chang Liu
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Nan Zhou
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Resources and Environmental Microbiology Research Center (EMRC), Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
- State Key Laboratory of Microbial Biotechnology, Shandong University, Qingdao 266237, PR China
- University of Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
30
|
Michael H, Amimo JO, Rajashekara G, Saif LJ, Vlasova AN. Mechanisms of Kwashiorkor-Associated Immune Suppression: Insights From Human, Mouse, and Pig Studies. Front Immunol 2022; 13:826268. [PMID: 35585989 PMCID: PMC9108366 DOI: 10.3389/fimmu.2022.826268] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/31/2022] [Indexed: 12/11/2022] Open
Abstract
Malnutrition refers to inadequate energy and/or nutrient intake. Malnutrition exhibits a bidirectional relationship with infections whereby malnutrition increases risk of infections that further aggravates malnutrition. Severe malnutrition (SM) is the main cause of secondary immune deficiency and mortality among children in developing countries. SM can manifest as marasmus (non-edematous), observed most often (68.6% of all malnutrition cases), kwashiorkor (edematous), detected in 23.8% of cases, and marasmic kwashiorkor, identified in ~7.6% of SM cases. Marasmus and kwashiorkor occur due to calorie-energy and protein-calorie deficiency (PCD), respectively. Kwashiorkor and marasmic kwashiorkor present with reduced protein levels, protein catabolism rates, and altered levels of micronutrients leading to uncontrolled oxidative stress, exhaustion of anaerobic commensals, and proliferation of pathobionts. Due to these alterations, kwashiorkor children present with profoundly impaired immune function, compromised intestinal barrier, and secondary micronutrient deficiencies. Kwashiorkor-induced alterations contribute to growth stunting and reduced efficacy of oral vaccines. SM is treated with antibiotics and ready-to-use therapeutic foods with variable efficacy. Kwashiorkor has been extensively investigated in gnotobiotic (Gn) mice and piglet models to understand its multiple immediate and long-term effects on children health. Due to numerous physiological and immunological similarities between pigs and humans, pig represents a highly relevant model to study kwashiorkor pathophysiology and immunology. Here we summarize the impact of kwashiorkor on children's health, immunity, and gut functions and review the relevant findings from human and animal studies. We also discuss the reciprocal interactions between PCD and rotavirus-a highly prevalent enteric childhood pathogen due to which pathogenesis and immunity are affected by childhood SM.
Collapse
Affiliation(s)
- Husheem Michael
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Joshua O. Amimo
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
- Department of Animal Production, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - Gireesh Rajashekara
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Linda J. Saif
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| | - Anastasia N. Vlasova
- Center for Food Animal Health, Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster, OH, United States
| |
Collapse
|
31
|
Xie T, Wang Y, Zou Z, Wu Y, Fan X, Dai J, Liu Y, Bai J. Relationship between the gut microbiota and temperament in children 1-2 years old in Chinese birth cohort. J Psychiatr Res 2022; 148:52-60. [PMID: 35101710 DOI: 10.1016/j.jpsychires.2022.01.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 12/07/2021] [Accepted: 01/17/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Understanding the relationship between the gut microbiota and temperament can provide new insights for the regulation of behavioral intervention in children, which is still lacking research. This study aimed to examine the relationship between the gut microbiota and temperament in a cohort of children in 1 year and 2 years old. METHODS This study included a total of 37 children with completed information, in which 51 samples at age 1 and 41 samples at age 2 were received respectively. We collected birth and demographic information. Parents reported their child's temperament characteristics using the Infant Behavior Questionnaire-revised (IBQ-R) and Early Childhood Behavior Questionnaire (ECBQ). Fecal samples were collected from each child at 1 and 2 years old and sequenced with MiSeq sequencer. Multiple linear regressions and linear mixed effect models were used to analyze the relationship between the temperament and their microbiota composition as well as the diversity and effect of gender or age on this relationship. RESULTS At age of year 2, Faecalibacterium was negatively associated with high-intensity pleasure and surgency. Bifidobacterium was negatively correlated with Perceptual sensitivity. Results showed no difference about three domains between year 1 and year 2, while gut microbiota showed diversity difference and genera difference. There was no gender and age difference on the relationship between temperament and the gut microbiota. CONCLUSIONS Temperament was associated with the gut microbiota over time. The temperament remained stable and the relationship between the gut microbiota and temperament wasn't associated with age and gender.
Collapse
Affiliation(s)
- Tianqu Xie
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
| | - Yuchen Wang
- Taiyuan Central Hospital of Shanxi Medical University, No. 5 Dong San Dao Xiang, Jiefang Road, Taiyuan, 030009, China.
| | - Zhijie Zou
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
| | - Yinyin Wu
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
| | - Xiaoxiao Fan
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
| | - Jiamiao Dai
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
| | - Yanqun Liu
- Wuhan University School of Nursing, Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
| | - Jinbing Bai
- Emory University Nell Hodgson Woodruff School of Nursing, 1520 Clifton Road, Atlanta, GA, 30322, USA.
| |
Collapse
|
32
|
Hu J, Zeng J, Shi Y, Song S. Are microbes and metabolites influencing the parental consumption of nestlings' feces in gray-backed shrikes? Curr Zool 2022; 68:667-678. [PMID: 36743228 PMCID: PMC9892794 DOI: 10.1093/cz/zoac005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/24/2022] [Indexed: 02/07/2023] Open
Abstract
The behavioral video recordings of the gray-backed shrike Lanius tephronotus revealed that parent birds eat the feces produced by their nestlings. "Parental nutrition hypothesis" attributes the origin of this behavior to nutrition-recovery and cost-saving, respectively. However, the presence of usable nutrients in the nestlings' feces is unknown because of traditional technology. In this study, we analyzed all the metabolites and the variations in the diversity and content of microbes in the feces of gray-backed shrike nestlings. We aimed to report the changes in microbes and metabolites with the age of nestlings and point out that the parent birds that eat the feces may gain potential nutrition benefits. The results showed that the relative abundances of Proteobacteria, Firmicutes, and Bacteroidota, changed significantly when the nestlings were 6 days old. The relative abundances of 6 probiotics, which are involved in digestion, metabolism, and immunity-related physiological functions, decreased in the nestlings' feces gradually with age; therefore, these probiotics may be obtained by parent birds upon ingestion of the feces of young nestlings. Among the metabolites that were detected, 20 were lipids and some had a role in anti-parasitic functions and wound healing; however, their relative contents decreased with age. These beneficial substances in the nestlings' feces may stimulate the parents to swallow the feces. Moreover, there were many aromatic metabolites in the newly hatched nestlings' feces, but the content of bitter metabolites increased as they grew up. Therefore, our results are in accordance with the nutritional hypothesis.
Collapse
Affiliation(s)
- Jie Hu
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jingyuan Zeng
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yurou Shi
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Sen Song
- Address correspondence to Sen Song. E-mail:
| |
Collapse
|
33
|
Sarr M, Tall ML, Ben Khedher M, Pham TPT, Mbaye B, Camara A, Armstrong N, Chartier C, Fadlane A, Sokhna C, Raoult D, Tidjani Alou M, Million M. Konateibacter massiliensis gen. nov. sp. nov. and Paenibacillus faecalis sp. nov., Two New Species Isolated from the Stool Samples of Infants Suffering from Marasmus. Curr Microbiol 2022; 79:68. [DOI: 10.1007/s00284-021-02757-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 12/30/2021] [Indexed: 11/28/2022]
|
34
|
Rubio-Del-Campo A, Gozalbo-Rovira R, Moya-Gonzálvez EM, Alberola J, Rodríguez-Díaz J, Yebra MJ. Infant gut microbiota modulation by human milk disaccharides in humanized microbiome mice. Gut Microbes 2022; 13:1-20. [PMID: 33938391 PMCID: PMC8096338 DOI: 10.1080/19490976.2021.1914377] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [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
Human milk glycans present a unique diversity of structures that suggest different mechanisms by which they may affect the infant microbiome development. A humanized mouse model generated by infant fecal transplantation was utilized here to evaluate the impact of fucosyl-α1,3-GlcNAc (3FN), fucosyl-α1,6-GlcNAc, lacto-N-biose (LNB) and galacto-N-biose on the fecal microbiota and host-microbiota interactions. 16S rRNA amplicon sequencing showed that certain bacterial genera significantly increased (Ruminococcus and Oscillospira) or decreased (Eubacterium and Clostridium) in all disaccharide-supplemented groups. Interestingly, cluster analysis differentiates the consumption of fucosyl-oligosaccharides from galactosyl-oligosaccharides, highlighting the disappearance of Akkermansia genus in both fucosyl-oligosaccharides. An increment of the relative abundance of Coprococcus genus was only observed with 3FN. As well, LNB significantly increased the relative abundance of Bifidobacterium, whereas the absolute levels of this genus, as measured by quantitative real-time PCR, did not significantly increase. OTUs corresponding to the species Bifidobacterium longum, Bifidobacterium adolescentis and Ruminococcus gnavus were not present in the control after the 3-week intervention, but were shared among the donor and specific disaccharide groups, indicating that their survival is dependent on disaccharide supplementation. The 3FN-feeding group showed increased levels of butyrate and acetate in the colon, and decreased levels of serum HDL-cholesterol. 3FN also down-regulated the pro-inflammatory cytokine TNF-α and up-regulated the anti-inflammatory cytokines IL-10 and IL-13, and the Toll-like receptor 2 in the large intestine tissue. The present study revealed that the four disaccharides show efficacy in producing beneficial compositional shifts of the gut microbiota and in addition, the 3FN demonstrated physiological and immunomodulatory roles.
Collapse
Affiliation(s)
- Antonio Rubio-Del-Campo
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, IATA-CSIC, Paterna, Spain
| | - Roberto Gozalbo-Rovira
- Departamento de Microbiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Eva M. Moya-Gonzálvez
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, IATA-CSIC, Paterna, Spain
| | - Juan Alberola
- Departamento de Microbiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Jesús Rodríguez-Díaz
- Departamento de Microbiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - María J. Yebra
- Laboratorio de Bacterias Lácticas y Probióticos, Departamento de Biotecnología de Alimentos, IATA-CSIC, Paterna, Spain,CONTACT María J. Yebra Laboratorio De Bacterias Lácticas Y Probióticos, Departamento De Biotecnología De Alimentos, IATA-CSIC, Agustín Escardino 7, 46980Paterna, Spain
| |
Collapse
|
35
|
Ahn JR, Lee SH, Kim B, Nam MH, Ahn YK, Park YM, Jeong SM, Park MJ, Song KB, Lee SY, Hong SJ. Ruminococcus gnavus ameliorates atopic dermatitis by enhancing Treg cell and metabolites in BALB/c mice. Pediatr Allergy Immunol 2022; 33:e13678. [PMID: 34633714 DOI: 10.1111/pai.13678] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Ruminococcus gnavus (R. gnavus) are mucin-degrading gut bacteria that play a key role in the early colonization of the gut by serving as endogenous sources of nutrients. They can also influence immune development. We had previously reported a lower abundance of R. gnavus in infants with atopic dermatitis (AD) compared with that in healthy subjects. However, the underlying mechanisms remain unclear. In this study, we investigated the effect of orally administered R. gnavus on antibiotic treatment-induced gut dysbiosis (and the underlying mechanism) in a mouse model of AD. METHODS Four-week-old female BALB/C mice were administered antibiotic cocktails for 2 weeks. R. gnavus was orally administered throughout the study duration. At 6 weeks of age, AD was induced by epidermal sensitization with ovalbumin. AD phenotypes and systemic and gut immune responses were investigated. RESULTS Orally administered R. gnavus significantly reduced AD-associated parameters (i.e., transepidermal water loss, clinical score, total serum immunoglobulin (Ig) E level, OVA-specific IgE level, and skin inflammation). R. gnavus treatment also resulted in significant downregulation of T helper 2-related cytokine mRNA and upregulation of interleukin (IL)-10 and Foxp3 in the skin. The population of CD4+ FOXP3+ T cells in mesenteric- and skin-draining lymph nodes and butyrate levels in the cecum increased in R. gnavus-administered AD mice. CONCLUSIONS Immune modulation by orally administered R. gnavus may alleviate AD symptoms through the enhancement of regulatory T-cell counts and short-chain fatty acids production in AD mice.
Collapse
Affiliation(s)
- Jae-Rin Ahn
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Seung-Hwa Lee
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Byunghyun Kim
- Korea Basic Science Institute, Seoul Center, Seoul, Korea
| | - Myung Hee Nam
- Korea Basic Science Institute, Seoul Center, Seoul, Korea
| | - Yoon Kyung Ahn
- Korea Basic Science Institute, Western Seoul Center, Seoul, Korea
| | - Yoon Mee Park
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Seon-Mi Jeong
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Korea
| | - Min Jee Park
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Kun Baek Song
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - So-Yeon Lee
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Soo-Jong Hong
- Department of Pediatrics, Childhood Asthma Atopy Center, Humidifier Disinfectant Health Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
36
|
Zhong N, Ma Y, Meng X, Sowanou A, Wu L, Huang W, Gao Y, Pei J. Effect of Fluoride in Drinking Water on Fecal Microbial Community in Rats. Biol Trace Elem Res 2022; 200:238-246. [PMID: 33576944 DOI: 10.1007/s12011-021-02617-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 01/27/2021] [Indexed: 12/16/2022]
Abstract
Intestinal nutrition has a close association with the onset and development of fluorosis. Intestinal microbes play a major role in intestinal nutrition. However, the effect of fluoride on intestinal microbes is still not fully understood. This study aimed to evaluate the dose-response of fluoride on fecal microbes as well as the link between fluorosis and fecal microbes. The results showed that fluoride did not significantly alter the diversity of fecal microbiota, but richness estimators (ACE and Chao) increased first, and then decreased with the increase of water fluoride. At the genus level, 150 mg/L fluoride significantly reduced the abundances of Roseburia and Clostridium sensu stricto, and 100 mg/L and 150 mg/L fluoride obviously increased the abundances of Unclassified Ruminococcaceaes and Unclassified Bdellovibrionales, respectively. The correlation analysis showed fluoride exposure had a negative association with Roseburia and Turicibacter and was positively associated with Pelagibacterium, Unclassified Ruminococcaceae, and Unclassified Bdellovibrionales. Dental fluorosis was negatively associated with Clostridium sensu stricto, Roseburia, Turicibacter, and Paenalcaligenes and had a positive association with Pelagibacterium, Unclassified Ruminococcaceae, and Unclassified Bdellovibrionales. In conclusion, this study firstly reports fluoride in drinking water has a remarkable biphasic effect on fecal microbiota in rats, and some bacteria are significantly associated with fluoride exposure and dental fluorosis. These results indicate the gut microbiota may play an important role in fluorosis, and some bacteria are likely to be developed as biomarkers for fluorosis.
Collapse
Affiliation(s)
- Nan Zhong
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission (23618504), Institute for Kaschin-Beck Disease Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Yongzheng Ma
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission (23618504), Institute for Kaschin-Beck Disease Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Xinyue Meng
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission (23618504), Institute for Kaschin-Beck Disease Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Alphonse Sowanou
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission (23618504), Institute for Kaschin-Beck Disease Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Liaowei Wu
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission (23618504), Institute for Fluorosis Disease Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Wei Huang
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission (23618504), Institute for Fluorosis Disease Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Yanhui Gao
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission (23618504), Institute for Fluorosis Disease Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Junrui Pei
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & National Health Commission (23618504), Institute for Kaschin-Beck Disease Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
37
|
Zhang L, Jia X, Lai P, Wang K, Bao Y, Li X. Relevance of Intestinal Microbiota in Immunoglobulin A Vasculitis With Abdominal Involvement. Front Pediatr 2022; 10:943267. [PMID: 35911834 PMCID: PMC9329519 DOI: 10.3389/fped.2022.943267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/21/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND We explored the intestinal microbiota changes in IgAV with abdominal involvement (IgAV-GI) at the acute and convalescent stages and evaluated the role of intestinal microbiota in the clinical course of patients with IgAV. METHODS A total of 37 patients with IgAV were included, and the control group comprised 37 age- and sex-matched healthy children. Stool samples were collected from 28 children with IgAV-GI (19 in the acute stage and 9 in the recovery stage) and from nine children with non-abdominal involvement. Fecal specimens were selected and DNA was obtained using an extraction kit which was then subjected to high-throughput sequencing and analysis. RESULTS There was no significant difference in the community structure of the intestinal microbiota among the IgAV-GI acute, IgAV-GI convalescence, and IgAV-non-GI stages. The abundance of Veillonella in the acute stage of IgAV-GI was significantly higher than that in IgAV-non-GI and convalescence stages, and Ruminococcus was the most abundant in IgAV-GI convalescence. The α-diversity of children with IgAV was significantly lower than that of healthy children, and healthy children had higher intestinal microbiota richness and more evenly distributed species. In terms of changes in intestinal microbial diversity in patients with IgAV at the genus level, obligate anaerobes such as Bifidobacterium, Prevotella, Coprobacter, Prevotella_9, Blautia, Romboutsia, Parabacteroide, Subdoligranulum, and Roseburia were significantly reduced, and the enrichment of facultative anaerobe was represented by Bacteroides, Lachnoclostridium, and Alistipe. CONCLUSION Different bacterial species may be involved in the pathogenesis of different types of IgAV-GI. Differences were observed in the intestinal microbiota between healthy children and children with IgAV.
Collapse
Affiliation(s)
- Linqian Zhang
- Jinhua Maternal and Child Health Care Hospital, Jinhua Women's and Children's Hospital, Jinhua, China
| | - Xinyi Jia
- Jinhua Maternal and Child Health Care Hospital, Jinhua Women's and Children's Hospital, Jinhua, China.,Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Panjian Lai
- Jinhua Maternal and Child Health Care Hospital, Jinhua Women's and Children's Hospital, Jinhua, China
| | - Kang Wang
- Jinhua Maternal and Child Health Care Hospital, Jinhua Women's and Children's Hospital, Jinhua, China
| | - Yunguang Bao
- Jinhua Maternal and Child Health Care Hospital, Jinhua Women's and Children's Hospital, Jinhua, China
| | - Xiaobing Li
- Jinhua Maternal and Child Health Care Hospital, Jinhua Women's and Children's Hospital, Jinhua, China
| |
Collapse
|
38
|
Coletto E, Latousakis D, Pontifex MG, Crost EH, Vaux L, Perez Santamarina E, Goldson A, Brion A, Hajihosseini MK, Vauzour D, Savva GM, Juge N. The role of the mucin-glycan foraging Ruminococcus gnavus in the communication between the gut and the brain. Gut Microbes 2022; 14:2073784. [PMID: 35579971 PMCID: PMC9122312 DOI: 10.1080/19490976.2022.2073784] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [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
Ruminococcus gnavus is a prevalent member of the human gut microbiota, which is over-represented in inflammatory bowel disease and neurological disorders. We previously showed that the ability of R. gnavus to forage on mucins is strain-dependent and associated with sialic acid metabolism. Here, we showed that mice monocolonized with R. gnavus ATCC 29149 (Rg-mice) display changes in major sialic acid derivatives in their cecum content, blood, and brain, which is accompanied by a significant decrease in the percentage of sialylated residues in intestinal mucins relative to germ-free (GF) mice. Changes in metabolites associated with brain function such as tryptamine, indolacetate, and trimethylamine N-oxide were also detected in the cecal content of Rg-mice when compared to GF mice. Next, we investigated the effect of R. gnavus monocolonization on hippocampus cell proliferation and behavior. We observed a significant decrease of PSA-NCAM immunoreactive granule cells in the dentate gyrus (DG) of Rg-mice as compared to GF mice and recruitment of phagocytic microglia in the vicinity. Behavioral assessments suggested an improvement of the spatial working memory in Rg-mice but no change in other cognitive functions. These results were also supported by a significant upregulation of genes involved in proliferation and neuroplasticity. Collectively, these data provide first insights into how R. gnavus metabolites may influence brain regulation and function through modulation of granule cell development and synaptic plasticity in the adult hippocampus. This work has implications for further understanding the mechanisms underpinning the role of R. gnavus in neurological disorders.
Collapse
Affiliation(s)
- Erika Coletto
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Dimitrios Latousakis
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Matthew G Pontifex
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Emmanuelle H Crost
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Laura Vaux
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Estella Perez Santamarina
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Andrew Goldson
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Arlaine Brion
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Mohammad K Hajihosseini
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - David Vauzour
- Norwich Medical School, Biomedical Research Centre, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - George M Savva
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| | - Nathalie Juge
- Gut Microbes and Health Institute Strategic Programme, Quadram Institute Bioscience, Norwich NR4 7UQ, UK
| |
Collapse
|
39
|
Wu H, Crost EH, Owen CD, van Bakel W, Martínez Gascueña A, Latousakis D, Hicks T, Walpole S, Urbanowicz PA, Ndeh D, Monaco S, Sánchez Salom L, Griffiths R, Reynolds RS, Colvile A, Spencer DIR, Walsh M, Angulo J, Juge N. The human gut symbiont Ruminococcus gnavus shows specificity to blood group A antigen during mucin glycan foraging: Implication for niche colonisation in the gastrointestinal tract. PLoS Biol 2021; 19:e3001498. [PMID: 34936658 PMCID: PMC8730463 DOI: 10.1371/journal.pbio.3001498] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 01/05/2022] [Accepted: 11/26/2021] [Indexed: 12/24/2022] Open
Abstract
The human gut symbiont Ruminococcus gnavus displays strain-specific repertoires of glycoside hydrolases (GHs) contributing to its spatial location in the gut. Sequence similarity network analysis identified strain-specific differences in blood-group endo-β-1,4-galactosidase belonging to the GH98 family. We determined the substrate and linkage specificities of GH98 from R. gnavus ATCC 29149, RgGH98, against a range of defined oligosaccharides and glycoconjugates including mucin. We showed by HPAEC-PAD and LC-FD-MS/MS that RgGH98 is specific for blood group A tetrasaccharide type II (BgA II). Isothermal titration calorimetry (ITC) and saturation transfer difference (STD) NMR confirmed RgGH98 affinity for blood group A over blood group B and H antigens. The molecular basis of RgGH98 strict specificity was further investigated using a combination of glycan microarrays, site-directed mutagenesis, and X-ray crystallography. The crystal structures of RgGH98 in complex with BgA trisaccharide (BgAtri) and of RgGH98 E411A with BgA II revealed a dedicated hydrogen network of residues, which were shown by site-directed mutagenesis to be critical to the recognition of the BgA epitope. We demonstrated experimentally that RgGH98 is part of an operon of 10 genes that is overexpresssed in vitro when R. gnavus ATCC 29149 is grown on mucin as sole carbon source as shown by RNAseq analysis and RT-qPCR confirmed RgGH98 expression on BgA II growth. Using MALDI-ToF MS, we showed that RgGH98 releases BgAtri from mucin and that pretreatment of mucin with RgGH98 confered R. gnavus E1 the ability to grow, by enabling the E1 strain to metabolise BgAtri and access the underlying mucin glycan chain. These data further support that the GH repertoire of R. gnavus strains enable them to colonise different nutritional niches in the human gut and has potential applications in diagnostic and therapeutics against infection.
Collapse
Affiliation(s)
- Haiyang Wu
- Quadram Institute Bioscience, Norwich, United Kingdom
| | | | - C David Owen
- Diamond Light Source Ltd, Didcot, United Kingdom
- Research Complex at Harwell, Didcot, United Kingdom
| | | | | | | | - Thomas Hicks
- University of East Anglia, Norwich, United Kingdom
| | | | | | - Didier Ndeh
- Quadram Institute Bioscience, Norwich, United Kingdom
| | | | | | | | | | - Anna Colvile
- Diamond Light Source Ltd, Didcot, United Kingdom
- Research Complex at Harwell, Didcot, United Kingdom
| | | | - Martin Walsh
- Diamond Light Source Ltd, Didcot, United Kingdom
- Research Complex at Harwell, Didcot, United Kingdom
| | - Jesus Angulo
- University of East Anglia, Norwich, United Kingdom
- Universidad de Sevilla and Instituto de Investigaciones Químicas, Sevilla, Spain
| | - Nathalie Juge
- Quadram Institute Bioscience, Norwich, United Kingdom
- * E-mail:
| |
Collapse
|
40
|
Assessing Gut Microbiota in an Infant with Congenital Propionic Acidemia before and after Probiotic Supplementation. Microorganisms 2021; 9:microorganisms9122599. [PMID: 34946200 PMCID: PMC8703847 DOI: 10.3390/microorganisms9122599] [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: 10/29/2021] [Revised: 11/25/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Propionic Acidemia (PA) is a rare inherited metabolic disorder caused by the enzymatic block of propionyl-CoA carboxylase with the consequent accumulation of propionic acid, which is toxic for the brain and cardiac cells. Since a considerable amount of propionate is produced by intestinal bacteria, interest arose in the attempt to reduce propionate-producing bacteria through a monthly antibiotic treatment of metronidazole. In the present study, we investigated the gut microbiota structure of an infant diagnosed at 4 days of life through Expanded Newborn Screening (NBS) and treated the child following international guidelines with a special low-protein diet, specific medications and strict biochemical monitoring. Microbiota composition was assessed during the first month of life, and the presence of Bacteroides fragilis, known to be associated with propionate production, was effectively decreased by metronidazole treatment. After five antibiotic therapy cycles, at 4 months of age, the infant was supplemented with a daily mixture of three bifidobacterial strains, known not to be propionate producers. The supplementation increased the population of bifidobacteria, with Bifidobacterium breve as the dominating species; Ruminococcus gnavus, an acetate and formate producer, was also identified. Metabarcoding analysis, compared with low coverage whole metagenome sequencing, proved to capture all the microbial biodiversity and could be the elected tool for fast and cost-effective monitoring protocols to be implemented in the follow up of rare metabolic disorders such as PA. Data obtained could be a possible starting point to set up tailored microbiota modification treatment studies in the attempt to improve the quality of life of people affected by propionic acidemia.
Collapse
|
41
|
Cui X, Wang Z, Tan Y, Chang S, Zheng H, Wang H, Yan T, Guru T, Hou F. Selenium Yeast Dietary Supplement Affects Rumen Bacterial Population Dynamics and Fermentation Parameters of Tibetan Sheep ( Ovis aries) in Alpine Meadow. Front Microbiol 2021; 12:663945. [PMID: 34276597 PMCID: PMC8283570 DOI: 10.3389/fmicb.2021.663945] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Selenium (Se) deficiency is a widespread and seasonally chronic phenomenon observed in Tibetan sheep (Ovis aries) traditionally grazed on the Qinghai-Tibet Plateau (QTP). Effects of the dietary addition of Se-enriched yeast (SeY) on the bacterial community in sheep rumen and rumen fermentation were evaluated with the aim of gaining a better understanding of the rumen prokaryotic community. Twenty-four yearling Tibetan rams [initial average body weight (BW) of 31.0 ± 0.64 kg] were randomly divided into four treatment groups, namely, control (CK), low Se (L), medium Se (M), and high Se (H). Each group comprised six rams and was fed a basic diet of fresh forage cut from the alpine meadow, to which SeY was added at prescribed dose rates. This feed trial was conducted for over 35 days. On the final day, rumen fluid was collected using a transesophageal sampler for analyzing rumen pH, NH3-N content, volatile fatty acid (VFA) level, and the rumen microbial community. Our analyses showed that NH3-N, total VFA, and propionate concentrations in the M group were significantly higher than in the other groups (P < 0.05). Both the principal coordinates analysis (PCoA) and the analysis of similarities revealed that the bacterial population structure of rumen differed among the four groups. The predominant rumen bacterial phyla were found to be Bacteroidetes and Firmicutes, and the three dominant genera in all the samples across all treatments were Christensenellaceae R7 group, Rikenellaceae RC9 gut group, and Prevotella 1. The relative abundances of Prevotella 1, Rikenellaceae RC9 gut group, Ruminococcus 2, Lachnospiraceae XPB1014 group, Carnobacterium, and Hafnia-Obesumbacterium were found to differ significantly among the four treatment groups (P < 0.05). Moreover, Tax4fun metagenome estimation revealed that gene functions and metabolic pathways associated with carbohydrate and other amino acids were overexpressed in the rumen microbiota of SeY-supplemented sheep. To conclude, SeY significantly affects the abundance of rumen bacteria and ultimately affects the rumen microbial fermentation.
Collapse
Affiliation(s)
- Xiongxiong Cui
- State Key Laboratory of Grassland Agro-ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Zhaofeng Wang
- State Key Laboratory of Grassland Agro-ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Yuhui Tan
- State Key Laboratory of Grassland Agro-ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Shenghua Chang
- State Key Laboratory of Grassland Agro-ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| | - Huiru Zheng
- School of Computing, Ulster University, Antrim, United Kingdom
| | - Haiying Wang
- School of Computing, Ulster University, Antrim, United Kingdom
| | - Tianhai Yan
- Sustainable Agri-Food Sciences Division, Agriculture Branch, Agri-Food and Biosciences Institute, Hillsborough, United Kingdom
| | - Tsedan Guru
- Animal Husbandry Science and Technology Demonstration Park of Maqu County, Gannan, China
| | - Fujiang Hou
- State Key Laboratory of Grassland Agro-ecosystems, Key Laboratory of Grassland Livestock Industry Innovation, Ministry of Agriculture and Rural Affairs, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, China
| |
Collapse
|
42
|
Vander Wyst KB, Ortega-Santos CP, Toffoli SN, Lahti CE, Whisner CM. Diet, adiposity, and the gut microbiota from infancy to adolescence: A systematic review. Obes Rev 2021; 22:e13175. [PMID: 33590719 PMCID: PMC10762698 DOI: 10.1111/obr.13175] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 12/14/2022]
Abstract
Early life gut microbiota are affected by several factors that make identification of microbial-adiposity relationships challenging. This review evaluates studies that have investigated the gut microbiota composition associated with adiposity in infants, children, and adolescents and provides evidence-based nutrition recommendations that address microbiota-adiposity links. Electronic databases were systematically searched through January 2020. Eligible studies were published in English and analyzed gut microbiota and adiposity among individuals aged birth to 18 years. Abstracts and full-text articles were reviewed by three independent reviewers. Of 45 full-text articles reviewed, 33 were included. No difference in abundance was found for Bacteroidetes (n = 7/15 articles), Firmicutes (n = 10/17), Actinobacteria (n = 8/12), Proteobacteria (n = 8/12), Tenericutes (n = 4/5), and Verrucomicrobia (n = 4/6) with adiposity. Lower abundance of Christensenellaceae (n = 3/5) and Rikenellaceae (n = 6/8) but higher abundance of F. prausnitzii (n = 3/5) and Prevotella (n = 5/7) were associated with adiposity. A lack of consensus exists for gut microbial composition associations with adiposity. A healthy gut microbiota is associated with a diet rich in fruits and vegetables with moderate consumption of animal fat and protein. Future research should use more robust sequencing technologies to identify all bacterial taxa associated with adiposity and evaluate how diet effects these adiposity-associated microbes.
Collapse
Affiliation(s)
- Kiley B Vander Wyst
- College of Health Solutions, Arizona State University, Phoenix, Arizona, USA
- Center for Health Promotion and Disease Prevention, Arizona State University, Phoenix, Arizona, USA
| | | | - Samantha N Toffoli
- College of Health Solutions, Arizona State University, Phoenix, Arizona, USA
| | - Caroline E Lahti
- College of Liberal Arts and Sciences, Arizona State University, Phoenix, Arizona, USA
| | - Corrie M Whisner
- College of Health Solutions, Arizona State University, Phoenix, Arizona, USA
| |
Collapse
|
43
|
Han C, Wu X, Zou N, Zhang Y, Yuan J, Gao Y, Chen W, Yao J, Li C, Hou J, Qin D. Cichorium pumilum Jacq Extract Inhibits LPS-Induced Inflammation via MAPK Signaling Pathway and Protects Rats From Hepatic Fibrosis Caused by Abnormalities in the Gut-Liver Axis. Front Pharmacol 2021; 12:683613. [PMID: 33995112 PMCID: PMC8117150 DOI: 10.3389/fphar.2021.683613] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 04/19/2021] [Indexed: 11/13/2022] Open
Abstract
The development of liver fibrosis is closely related to the gut microbiota, and the "gut-liver axis" is the most important connection between the two. ethyl acetate extract of Cichorium pumilum Jacq (CGEA) is an herbal extract consisting mainly of sesquiterpenoids. The anti-inflammatory and hepatoprotective effects of CGEA have been reported, but the anti-fibrotic effects of CGEA via intestinal microbes and the "gut-liver axis" cycle have rarely been reported. In this study, we observed that CGEA not only directly attenuated inflammatory factor levels in inflamed mice, but also attenuated liver inflammation as well as liver fibrosis degeneration in rats with liver fibrosis caused by colitis. We observed in vitro that CGEA significantly promoted the growth of Bifidobacterium adolescentis. Similarly, fecal 16S rDNA sequencing of liver fibrosis rats showed that CGEA intervention significantly altered the composition of the intestinal microbiota of liver fibrosis rats. CGEA increased the abundance of intestinal microbiota, specifically, CGEA increased the ratio of Firmicutes to Bacteroidetes, CGEA could significantly increase the levels of Ruminococcus. In addition, CGEA intervention significantly protected intestinal mucosal tissues and improved intestinal barrier function in rats. Lactucin is the main sesquiterpenoid in CGEA, and HPLC results showed its content in CGEA was up to 6%. Lactucin has been reported to have significant anti-inflammatory activity, and in this study, we found that Lactucin decreased p38 kinases (p38), phosphorylation of the extracellular signal-regulated kinase (ERK) and protein kinase B (AKT) protein phosphorylation in lipopolysaccharide (LPS)-activated RAW264.7 cells, thereby reducing mRNA expression and protein expression of pro-inflammatory factors inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2), and inhibiting the release of inflammatory factors interleukin (IL)-6 and nitric oxide (NO), exerting anti-inflammatory effects. In summary, the prevention of liver fibrosis caused by intestinal inflammation by CGEA may be achieved by regulating the intestinal microbiota and restoring the intestinal barrier thereby improving the "gut-liver axis" circulation, reducing liver inflammation, and ultimately alleviating liver fibrosis. Notably, the direct anti-inflammatory effect of CGEA may be due to its content of Lactucin, which can exert anti-inflammatory effects by inhibiting the phosphorylation of Mitogen-activated protein kinase (MAPK) and Akt signaling pathways.
Collapse
Affiliation(s)
- Chang Han
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| | - Xi Wu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Nan Zou
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Yunsheng Zhang
- Husbandry Research Institute, Xinjiang Academy of Animal Science, Urumqi, China
| | - Jinqi Yuan
- First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Yuefeng Gao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Wen Chen
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| | - Jia Yao
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| | - Cong Li
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| | - Jinqiu Hou
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| | - Dongmei Qin
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| |
Collapse
|
44
|
Raspini B, Vacca M, Porri D, De Giuseppe R, Calabrese FM, Chieppa M, Liso M, Cerbo RM, Civardi E, Garofoli F, De Angelis M, Cena H. Early Life Microbiota Colonization at Six Months of Age: A Transitional Time Point. Front Cell Infect Microbiol 2021; 11:590202. [PMID: 33842380 PMCID: PMC8032992 DOI: 10.3389/fcimb.2021.590202] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
Background Early life gut microbiota is involved in several biological processes, particularly metabolism, immunity, and cognitive neurodevelopment. Perturbation in the infant’s gut microbiota increases the risk for diseases in early and later life, highlighting the importance of understanding the connections between perinatal factors with early life microbial composition. The present research paper is aimed at exploring the prenatal and postnatal factors influencing the infant gut microbiota composition at six months of age. Methods Gut microbiota of infants enrolled in the longitudinal, prospective, observational study “A.MA.MI” (Alimentazione MAmma e bambino nei primi MIlle giorni) was analyzed. We collected and analyzed 61 fecal samples at baseline (meconium, T0); at six months of age (T2), we collected and analyzed 53 fecal samples. Samples were grouped based on maternal and gestational weight factors, type of delivery, type of feeding, time of weaning, and presence/absence of older siblings. Alpha and beta diversities were evaluated to describe microbiota composition. Multivariate analyses were performed to understand the impact of the aforementioned factors on the infant’s microbiota composition at six months of age. Results Different clustering hypotheses have been tested to evaluate the impact of known metadata factors on the infant microbiota. Neither maternal body mass index nor gestational weight gain was able to determine significant differences in infant microbiota composition six months of age. Concerning the type of feeding, we observed a low alpha diversity in exclusive breastfed infants; conversely, non-exclusively breastfed infants reported an overgrowth of Ruminococcaceae and Flavonifractor. Furthermore, we did not find any statistically significant difference resulting from an early introduction of solid foods (before 4 months of age). Lastly, our sample showed a higher abundance of clostridial patterns in firstborn babies when compared to infants with older siblings in the family. Conclusion Our findings showed that, at this stage of life, there is not a single factor able to affect in a distinct way the infants’ gut microbiota development. Rather, there seems to be a complex multifactorial interaction between maternal and neonatal factors determining a unique microbial niche in the gastrointestinal tract.
Collapse
Affiliation(s)
- Benedetta Raspini
- Department of Public Health, Experimental and Forensic Medicine, Dietetics and Clinical Nutrition Laboratory, University of Pavia, Pavia, Italy
| | - Mirco Vacca
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Debora Porri
- Department of Public Health, Experimental and Forensic Medicine, Dietetics and Clinical Nutrition Laboratory, University of Pavia, Pavia, Italy
| | - Rachele De Giuseppe
- Department of Public Health, Experimental and Forensic Medicine, Dietetics and Clinical Nutrition Laboratory, University of Pavia, Pavia, Italy
| | | | - Marcello Chieppa
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, Castellana Grotte, Italy
| | - Marina Liso
- National Institute of Gastroenterology "S. de Bellis", Institute of Research, Castellana Grotte, Italy
| | - Rosa Maria Cerbo
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elisa Civardi
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Francesca Garofoli
- Neonatal Unit and Neonatal Intensive Care Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Maria De Angelis
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Hellas Cena
- Department of Public Health, Experimental and Forensic Medicine, Dietetics and Clinical Nutrition Laboratory, University of Pavia, Pavia, Italy.,Clinical Nutrition and Dietetics Service, Unit of Internal Medicine and Endocrinology, ICS Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
45
|
Interaction of Intestinal Bacteria with Human Rotavirus during Infection in Children. Int J Mol Sci 2021; 22:ijms22031010. [PMID: 33498321 PMCID: PMC7864024 DOI: 10.3390/ijms22031010] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/30/2022] Open
Abstract
The gut microbiota has emerged as a key factor in the pathogenesis of intestinal viruses, including enteroviruses, noroviruses and rotaviruses (RVs), where stimulatory and inhibitory effects on infectivity have been reported. With the aim of determining whether members of the microbiota interact with RVs during infection, a combination of anti-RV antibody labeling, fluorescence-activated cell sorting and 16S rRNA amplicon sequencing was used to characterize the interaction between specific bacteria and RV in stool samples of children suffering from diarrhea produced by G1P[8] RV. The genera Ruminococcus and Oxalobacter were identified as RV binders in stools, displaying enrichments between 4.8- and 5.4-fold compared to samples nonlabeled with anti-RV antibodies. In vitro binding of the G1P[8] Wa human RV strain to two Ruminococcus gauvreauii human isolates was confirmed by fluorescence microscopy. Analysis in R. gauvreauii with antibodies directed to several histo-blood group antigens (HBGAs) indicated that these bacteria express HBGA-like substances on their surfaces, which can be the target for RV binding. Furthermore, in vitro infection of the Wa strain in differentiated Caco-2 cells was significantly reduced by incubation with R. gauvreauii. These data, together with previous findings showing a negative correlation between Ruminococcus levels and antibody titers to RV in healthy individuals, suggest a pivotal interaction between this bacterial group and human RV. These results reveal likely mechanisms of how specific bacterial taxa of the intestinal microbiota could negatively affect RV infection and open new possibilities for antiviral strategies.
Collapse
|
46
|
Inhibition of PD-1 Protects against TNBS-Induced Colitis via Alteration of Enteric Microbiota. BIOMED RESEARCH INTERNATIONAL 2021; 2021:4192451. [PMID: 33506015 PMCID: PMC7810563 DOI: 10.1155/2021/4192451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 11/14/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022]
Abstract
Methods Colitis was induced in mice using 2,4,6-trinitrobenzene-sulfonic acid (TNBS), and mice were subsequently treated with either a PD-1 inhibitor or 5-amino-salicylic acid (ASA) as a positive control. Body weight, disease activity index (DAI), colon length, and tissue damage were evaluated, and the enteric microbiota was profiled using high-throughput 16S rRNA sequencing of fecal samples from the experimental mice. Results TNBS caused mice to experience IBD-like symptoms, which were attenuated by the PD-1 inhibitor, as indicated by a decrease in DAI scores (p = 0.0002). Furthermore, in this mouse model of IBD, PD-1 inhibition improved the alpha diversity as well as restored the beta diversity of the enteric microbiome. It also significantly enriched the abundance of short-chain fatty acid- (SCFA-) producing bacteria of the Firmicutes (p < 0.05) and Bacteroidetes (p < 0.05) phyla but depopulated Proteobacteria (p < 0.05). Conclusion PD-1 inhibition can partly mitigate TNBS-induced colitis and restore the enteric microbiota by enriching the abundance of SCFA-producing bacteria.
Collapse
|
47
|
Luise D, Spinelli E, Correa F, Nicodemo A, Bosi P, Trevisi P. The effect of a single, early-life administration of a probiotic on piglet growth performance and faecal microbiota until weaning. ITALIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1080/1828051x.2021.1952909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Diana Luise
- Department of Agricultural and Food Science (DISTAL), University of Bologna, Bologna, Italy
| | - Elisa Spinelli
- Department of Agricultural and Food Science (DISTAL), University of Bologna, Bologna, Italy
| | - Federico Correa
- Department of Agricultural and Food Science (DISTAL), University of Bologna, Bologna, Italy
| | | | - Paolo Bosi
- Department of Agricultural and Food Science (DISTAL), University of Bologna, Bologna, Italy
| | - Paolo Trevisi
- Department of Agricultural and Food Science (DISTAL), University of Bologna, Bologna, Italy
| |
Collapse
|
48
|
Egan M, Dempsey E, Ryan CA, Ross RP, Stanton C. The Sporobiota of the Human Gut. Gut Microbes 2021; 13:1-17. [PMID: 33406976 PMCID: PMC7801112 DOI: 10.1080/19490976.2020.1863134] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 02/04/2023] Open
Abstract
The human gut microbiome is a diverse and complex ecosystem that plays a critical role in health and disease. The composition of the gut microbiome has been well studied across all stages of life. In recent years, studies have investigated the production of endospores by specific members of the gut microbiome. An endospore is a tough, dormant structure formed by members of the Firmicutes phylum, which allows for greater resistance to otherwise inhospitable conditions. This innate resistance has consequences for human health and disease, as well as in biotechnology. In particular, the formation of endospores is strongly linked to antibiotic resistance and the spread of antibiotic resistance genes, also known as the resistome. The term sporobiota has been used to define the spore-forming cohort of a microbial community. In this review, we present an overview of the current knowledge of the sporobiota in the human gut. We discuss the development of the sporobiota in the infant gut and the perinatal factors that may have an effect on vertical transmission from mother to infant. Finally, we examine the sporobiota of critically important food sources for the developing infant, breast milk and powdered infant formula.
Collapse
Affiliation(s)
- Muireann Egan
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Eugene Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - C. Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
49
|
Alsharairi NA. The Role of Short-Chain Fatty Acids in the Interplay between a Very Low-Calorie Ketogenic Diet and the Infant Gut Microbiota and Its Therapeutic Implications for Reducing Asthma. Int J Mol Sci 2020; 21:E9580. [PMID: 33339172 PMCID: PMC7765661 DOI: 10.3390/ijms21249580] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/14/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota is well known as playing a critical role in inflammation and asthma development. The very low-calorie ketogenic diet (VLCKD) is suggested to affect gut microbiota; however, the effects of VLCKD during pregnancy and lactation on the infant gut microbiota are unclear. The VLCKD appears to be more effective than caloric/energy restriction diets for the treatment of several diseases, such as obesity and diabetes. However, whether adherence to VLCKD affects the infant gut microbiota and the protective effects thereof on asthma remains uncertain. The exact mechanisms underlying this process, and in particular the potential role of short chain fatty acids (SCFAs), are still to be unravelled. Thus, the aim of this review is to identify the potential role of SCFAs that underlie the effects of VLCKD during pregnancy and lactation on the infant gut microbiota, and explore whether it incurs significant implications for reducing asthma.
Collapse
Affiliation(s)
- Naser A Alsharairi
- Heart, Mind & Body Research Group, Menzies Health Institute Queensland, Griffith University, Gold Coast 4222, Australia
| |
Collapse
|
50
|
Te Pas MFW, Jansman AJM, Kruijt L, van der Meer Y, Vervoort JJM, Schokker D. Sanitary Conditions Affect the Colonic Microbiome and the Colonic and Systemic Metabolome of Female Pigs. Front Vet Sci 2020; 7:585730. [PMID: 33195612 PMCID: PMC7649119 DOI: 10.3389/fvets.2020.585730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/22/2020] [Indexed: 12/04/2022] Open
Abstract
Differences in sanitary conditions, as model to induce differences in subclinical immune stimulation, affect the growth performance and nutrient metabolism in pigs. The objective of the present study was to evaluate the colonic microbiota and the colonic and systemic metabolome of female pigs differing in health status induced by sanitary conditions. We analyzed blood and colon digesta metabolite profiles using Nuclear Magnetic Resonance (1H NMR) and Triple quadrupole mass spectrometry, as well as colonic microbiota profiles. 1H NMR is a quantitative metabolomics technique applicable to biological samples. Weaned piglets of 4 weeks of age were kept under high or low sanitary conditions for the first 9 weeks of life. The microbiota diversity in colon digesta was higher in pigs subjected to low sanitary conditions (n = 18 per treatment group). The abundance of 34 bacterial genera was higher in colon digesta of low sanitary condition pigs, while colon digesta of high sanitary status pigs showed a higher abundance for four bacterial groups including the Megasphaera genus (p < 0.003) involved in lactate fermentation. Metabolite profiles (n = 18 per treatment group) in blood were different between both groups of pigs. These different profiles suggested changes in general nutrient metabolism, and more specifically in amino acid metabolism. Moreover, differences in compounds related to the immune system and responses to stress were observed. Microbiome-specific metabolites in blood were also affected by sanitary status of the pigs. We conclude that the microbiome composition in colon and the systemic metabolite profiles are affected by sanitary conditions and related to suboptimal health. These data are useful for exploring further relationships between health, metabolic status and performance and for the identification of biomarkers related to health (indices) and performance.
Collapse
Affiliation(s)
- Marinus F W Te Pas
- Wageningen Livestock Research, Wageningen University and Research, Wageningen, Netherlands
| | - Alfons J M Jansman
- Wageningen Livestock Research, Wageningen University and Research, Wageningen, Netherlands
| | - Leo Kruijt
- Wageningen Livestock Research, Wageningen University and Research, Wageningen, Netherlands
| | - Yvonne van der Meer
- Wageningen Livestock Research, Wageningen University and Research, Wageningen, Netherlands
| | - Jacques J M Vervoort
- Department of Agrotechnology and Food Sciences, Biochemistry, Wageningen University, Wageningen, Netherlands
| | - Dirkjan Schokker
- Wageningen Livestock Research, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|