1
|
Yang L, Yao B, Zhang S, Yang Y, Wang G, Pan H, Zeng X, Qiao S. Division mechanism of labor in Diqing Tibetan Pigs gut microbiota for dietary fiber efficiently utilization. Microbiol Res 2025; 290:127977. [PMID: 39577368 DOI: 10.1016/j.micres.2024.127977] [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: 08/26/2024] [Revised: 10/25/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024]
Abstract
The Diqing Tibetan (TP) pig is an roughage tolerance breed that inhabits an area with the highest altitude distribution in the world and can be maintained on a diet containing 90 % forage material in confined production systems. Our results showed that TP pigs had a strong capability for high-efficiency utilization of arabinose and xylose. Metagenomic analysis revealed that the secretion of carbohydrate esterases was mainly undertaken by fecal strains of Microbacterium, Alistipes, Acinetobacter, and Faecalibacterium, while Microbacterium, Prevotella, Turicibacter, Lactobacillus, Clostridium, and Streptococcus were responsible for most of the secretion of glycoside hydrolases. Then, a brand new species, which was named Microbacterium sp. Qiao 01 was captured and appeared to have the highest fiber utilization ability in vitro, degrading 36.54 % of the neutral detergent fiber in corn stover. Our results provide strong evidence that efficient utilization of dietary fiber by TP pigs is due to the emergence of highly specialized microbial strategies in the gut. Microorganisms showed preferences and a clear division of labor in the degradation process of dietary fiber. This study has great practical significance for improving the utilization efficiency of livestock feed and alleviating the tension of food insecurity.
Collapse
Affiliation(s)
- Lijie Yang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2, Haidian District, Beijing 100193, China; Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Shandong Provincial Key Laboratory of Animal Nutrition and Efficient Feeding,Department of Animal Science, Shandong Agricultural University, Tai'an, Shandong 271017, China.
| | - Bingqian Yao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2, Haidian District, Beijing 100193, China.
| | - Shimin Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2, Haidian District, Beijing 100193, China.
| | - Yuting Yang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2, Haidian District, Beijing 100193, China.
| | - Gang Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2, Haidian District, Beijing 100193, China.
| | - Hongbin Pan
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2, Haidian District, Beijing 100193, China.
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2, Haidian District, Beijing 100193, China.
| |
Collapse
|
2
|
Vásquez-Reyes S, Bernal-Gámez M, Domínguez-Chávez J, Mondragón-Vásquez K, Sánchez-Tapia M, Ordaz G, Granados-Portillo O, Coutiño-Hernández D, Barrera-Gómez P, Torres N, Tovar AR. The Effects of Novel Co-Amorphous Naringenin and Fisetin Compounds on a Diet-Induced Obesity Murine Model. Nutrients 2024; 16:4425. [PMID: 39771046 PMCID: PMC11678459 DOI: 10.3390/nu16244425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVE In recent studies, it has been shown that dietary bioactive compounds can produce health benefits; however, it is not known whether an improvement in solubility can enhance their biological effects. Thus, the aim of this work was to study whether co-amorphous (CoA) naringenin or fisetin with enhanced solubility modify glucose and lipid metabolism, thermogenic capacity and gut microbiota in mice fed a high-fat, high-sucrose (HFSD) diet. METHODS Mice were fed with an HFSD with or without CoA-naringenin or CoA-fisetin for 3 months. Body weight, food intake, body composition, glucose tolerance, hepatic lipid composition and gut microbiota were assessed. RESULTS CoA-naringenin demonstrated significant reductions in fat-mass gain, improved cholesterol metabolism, and enhanced glucose tolerance. Mice treated with CoA-naringenin gained 45% less fat mass and exhibited improved hepatic lipid profiles, with significant reductions seen in liver triglycerides and cholesterol. Additionally, both CoA-flavonoids increased oxygen consumption (VO2), contributing to enhanced energy expenditure and improved metabolic flexibility. Thermogenic activation, indicated by increased UCP1 and PGC-1α levels, was observed with CoA-fisetin, supporting its role in fat oxidation and adipocyte size reduction. Further, both CoA-flavonoids modulated gut microbiota, restoring diversity and promoting beneficial bacteria, such as Akkermansia muciniphila, which has been linked to improved metabolic health. CONCLUSIONS These findings suggest that co-amorphous naringenin or fisetin offers promising applications in improving solubility, metabolic health, and thermogenesis, highlighting the potential of both as therapeutic agents against obesity and related disorders.
Collapse
Affiliation(s)
- Sarai Vásquez-Reyes
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, México 14080, Mexico; (S.V.-R.); (M.B.-G.); (M.S.-T.); (G.O.); (O.G.-P.); (D.C.-H.); (P.B.-G.); (N.T.)
| | - Miranda Bernal-Gámez
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, México 14080, Mexico; (S.V.-R.); (M.B.-G.); (M.S.-T.); (G.O.); (O.G.-P.); (D.C.-H.); (P.B.-G.); (N.T.)
| | - Jorge Domínguez-Chávez
- Facultad de Bioanálisis Región Veracruz, Universidad Veracruzana, Agustín de Iturbide, Veracruz 91700, Mexico; (J.D.-C.); (K.M.-V.)
| | - Karina Mondragón-Vásquez
- Facultad de Bioanálisis Región Veracruz, Universidad Veracruzana, Agustín de Iturbide, Veracruz 91700, Mexico; (J.D.-C.); (K.M.-V.)
| | - Mónica Sánchez-Tapia
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, México 14080, Mexico; (S.V.-R.); (M.B.-G.); (M.S.-T.); (G.O.); (O.G.-P.); (D.C.-H.); (P.B.-G.); (N.T.)
| | - Guillermo Ordaz
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, México 14080, Mexico; (S.V.-R.); (M.B.-G.); (M.S.-T.); (G.O.); (O.G.-P.); (D.C.-H.); (P.B.-G.); (N.T.)
| | - Omar Granados-Portillo
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, México 14080, Mexico; (S.V.-R.); (M.B.-G.); (M.S.-T.); (G.O.); (O.G.-P.); (D.C.-H.); (P.B.-G.); (N.T.)
| | - Diana Coutiño-Hernández
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, México 14080, Mexico; (S.V.-R.); (M.B.-G.); (M.S.-T.); (G.O.); (O.G.-P.); (D.C.-H.); (P.B.-G.); (N.T.)
| | - Paulina Barrera-Gómez
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, México 14080, Mexico; (S.V.-R.); (M.B.-G.); (M.S.-T.); (G.O.); (O.G.-P.); (D.C.-H.); (P.B.-G.); (N.T.)
| | - Nimbe Torres
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, México 14080, Mexico; (S.V.-R.); (M.B.-G.); (M.S.-T.); (G.O.); (O.G.-P.); (D.C.-H.); (P.B.-G.); (N.T.)
| | - Armando R. Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, CDMX, México 14080, Mexico; (S.V.-R.); (M.B.-G.); (M.S.-T.); (G.O.); (O.G.-P.); (D.C.-H.); (P.B.-G.); (N.T.)
| |
Collapse
|
3
|
Gao F, Shen Y, Wu H, Laue HE, Lau FK, Gillet V, Lai Y, Shrubsole MJ, Prada D, Zhang W, Liu Z, Bellenger JP, Takser L, Baccarelli AA. Associations of Stool Metal Exposures with Childhood Gut Microbiome Multiomics Profiles in a Prospective Birth Cohort Study. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:22053-22063. [PMID: 39630952 DOI: 10.1021/acs.est.4c09642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Metal exposures are closely related to childhood developmental health. However, their effects on the childhood gut microbiome, which also impacts health, are largely unexplored using microbiome multiomics including the metagenome and metatranscriptome. This study examined the associations of fecal profiles of metal/element exposures with gut microbiome species and active functional pathways in 8- to 12-year-old children (N = 116) participating in the GESTation and Environment (GESTE) cohort study. We analyzed 19 stool metal and element concentrations (B, Na, Mg, Al, K, Ca, V, Cr, Mn, Fe, Co, Ni, Cu, Zn, As, Mo, Cd, Ba, and Pb). Covariate-adjusted linear regression models identified several significant microbiome associations with continuous stool metal/element concentrations. For instance, Zn was positively associated with Turicibacter sanguinis (coef = 1.354, q-value = 0.039) and negatively associated with Eubacterium eligens (coef = -0.794, q-value = 0.044). Higher concentrations of Cd were associated with lower Eubacterium eligens (coef = -0.774, q-value = 0.045). Additionally, a total of 490 significant functional pathways such as biosynthesis and degradation/utilization/assimilation were identified, corresponding to different functions, including amino acid synthesis and carbohydrate degradation. Our results suggest links among metal exposures, pediatric gut microbiome multiomics, and potential health implications. Future work will further explore their relation to childhood health.
Collapse
Affiliation(s)
- Feng Gao
- Department of Environmental Health Sciences, Fielding School of Public Health, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California 90095, United States
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Yike Shen
- Department of Earth and Environmental Sciences, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Haotian Wu
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Hannah E Laue
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst School of Public Health and Health Sciences, Amherst, Massachusetts 01003, United States
| | - Fion K Lau
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Virginie Gillet
- Département de Pédiatrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Yunjia Lai
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York 10032, United States
| | - Martha J Shrubsole
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Diddier Prada
- Institute for Health Equity Research - IHER, Department of Population Health Science and Policy and the Department of Environmental Medicine and Climate Science, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Wei Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Zhonghua Liu
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, New York 10032, United States
| | | | - Larissa Takser
- Département de Pédiatrie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec J1H 5N4, Canada
| | - Andrea A Baccarelli
- Office of the Dean, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, United States
| |
Collapse
|
4
|
Wang Z, Gao B, Liu X, Li A. The mediating role of metabolites between gut microbiome and Hirschsprung disease: a bidirectional two-step Mendelian randomization study. Front Pediatr 2024; 12:1371933. [PMID: 39258147 PMCID: PMC11384983 DOI: 10.3389/fped.2024.1371933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024] Open
Abstract
Background Gut microbiome (GM) was observed to be associated with the incidence of Hirschsprung disease (HD). However, the effect and mechanism of GM in HD is still unclear. To investigate the relationship between GM and HD and the effect of metabolites as mediators, a bidirectional two-step Mendelian randomization (MR) study was conducted. Methods The study selected instrument variables (IVs) from summary-level genome-wide association studies (GWAS). The MiBioGen consortium provided the GWAS data for GM, while the GWAS data for metabolites and HD were obtained from the GWAS Catalog consortium. Two-sample MR analyses were performed to estimate bidirectional correlations between IVs associated with GM and HD. Then, genetic variants related to 1,400 metabolite traits were selected for further mediation analyses using the Product method. Results This study found that seven genus bacteria had a significant causal relationship with the incidence of HD but not vice versa. 27 metabolite traits were significantly correlated with HD. After combining the significant results, three significant GM-metabolites-HD lines have been identified. In the Peptococcus-Stearoyl sphingomyelin (d18:1/18:0)-HD line, the Stearoyl sphingomyelin (d18:1/18:0) levels showed a mediation proportion of 14.5%, while in the Peptococcus-lysine-HD line, the lysine levels had a mediation proportion of 12.9%. Additionally, in the Roseburia-X-21733-HD line, the X-21733 levels played a mediation proportion of 23.5%. Conclusion Our MR study indicates a protective effect of Peptococcus on HD risk that is partially mediated through serum levels of stearoyl sphingomyelin (d18:1/18:0) and lysine, and a risk effect of Roseburia on HD that is partially mediated by X-21733 levels. These findings could serve as novel biomarkers and therapeutic targets for HD.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Bingjun Gao
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao Liu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, China
| | - Aiwu Li
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
5
|
Chong CW, Liew MS, Ooi W, Jamil H, Lim A, Hooi SL, Tay CSC, Tan G. Effect of green banana and pineapple fibre powder consumption on host gut microbiome. Front Nutr 2024; 11:1437645. [PMID: 39246394 PMCID: PMC11378528 DOI: 10.3389/fnut.2024.1437645] [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: 05/24/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Purpose To determine whether green banana powder (GBP) and pineapple fibre powder (PFP) promote beneficial bacterial species, directly improve human gut health and modulate the gut microbiome and understand their utility as functional foods and dietary supplements. Methods Over 14 days, 60 adults followed protocol requirements, completed food diaries and study questionnaires, avoided consuming supplements with prebiotics, probiotics or postbiotics, and ingested food containing 5 g of total daily fibre [placebo (10.75 g), GBP (10.75 g) or PFP (7.41 g)]. Participants' medical and baseline wellness histories, as well as stool samples, were collected at baseline, day 7 and 14. Stool DNA was processed for sequencing. Results Dietary fibre and resistant starches (RS) in GBP and PFP promoted temporal increases in beneficial bacteria. GBP significantly elevated 7 species (F. prausnitzii, B. longum, B. bifidum, B. adolescentis, B. pseudocatenulatum, B. obeum, and R. inulinivorans), while PFP enriched 6 species (B. ovatus, B. cellulosilyticus, B. bifidum, B. intestinalis, R. inulinivorans, and E. siraeum). These bacteria, found to be deficient in younger adults, were promoted by both powders. PFP benefitted both genders aged 16-23, while GBP benefitted overweight/obese individuals, including females. GBP and PFP fiber and RS improved bowel regularity and health as well as metabolism by promoting histidine, branched-chain amino acids, short-chain fatty acids, and biotin production. The additional fiber caused "low" bloatedness and reduced "fairly bad" sleep disruptions, without affecting sleep durations. Conclusion GBP and PFP supplementation increased beneficial bacteria and metabolites, improved host gut health, and present a valuable nutritional strategy for enhancing human health. Clinical trial registration AMILI Institutional Review Board, Identifier 2023/0301.
Collapse
Affiliation(s)
- Chun Wie Chong
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
| | - Mei Shan Liew
- Dole Specialty Ingredients, Dole Asia Holdings Pte., Ltd., Singapore, Singapore
| | - Weitze Ooi
- Dole Specialty Ingredients, Dole Asia Holdings Pte., Ltd., Singapore, Singapore
| | - Hassan Jamil
- Dole Specialty Ingredients, Dole Asia Holdings Pte., Ltd., Singapore, Singapore
| | | | | | | | | |
Collapse
|
6
|
Fedorin MM, Livzan MA, Gaus OV, Pashkova EV. Potential role of short-chain fatty acids in irritable bowel syndrome in overweight and obese individuals. MEDITSINSKIY SOVET = MEDICAL COUNCIL 2024:20-27. [DOI: 10.21518/ms2024-168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
Due to increasing prevalence of functional diseases of the colon in obese patients, the mechanisms by which the intestinal microbiota affects the development of symptoms of irritable bowel syndrome (IBS) in the setting of metabolic activity of adipose tissue should be investigated. The quantitative and qualitative changes in the pool of synthesized short-chain fatty acids, which have a multidirectional impact on the colonic motility is one of the key mechanisms by which the intestinal microbiota affects the occurrence and features of the course of irritable bowel syndrome. But as regards the issue of whether individual short-chain fatty acids have an impact on the severity of abdominal pain and characteristics of colonic motility dysfunction, it remains a subject of discussions. The study of the mechanisms of impact of short-chain fatty acids on the development and progression of obesity deserves special attention. Increased serum and faecal short-chain fatty acid levels in obese patients can either be a result of changes in the intestinal microflora composition associated with special eating habits and lifestyle, or have an independent effect on the development of obesity in individuals due to intestinal microflora composition disorders that have been already developed. Due to special features of the course of irritable bowel syndrome associated with overweight and obesity, studying the intestinal microbiota composition and the short-chain fatty acids produced by it in this cohort of IBS patients is of particular interest. This publication has been prepared to describe and systematize the possible mechanisms of impact of short-chain fatty acids on the development of abdominal pain and impaired colonic motility in IBS patients with overweight and obesity. The literature search was conducted in the databases Embase, PubMed and Google Scholar using the keywords “irritable bowel syndrome”, “obesity”, “short-chain fatty acids”, “gut microbiota”.
Collapse
|
7
|
Mohr AE, Sweazea KL, Bowes DA, Jasbi P, Whisner CM, Sears DD, Krajmalnik-Brown R, Jin Y, Gu H, Klein-Seetharaman J, Arciero KM, Gumpricht E, Arciero PJ. Gut microbiome remodeling and metabolomic profile improves in response to protein pacing with intermittent fasting versus continuous caloric restriction. Nat Commun 2024; 15:4155. [PMID: 38806467 PMCID: PMC11133430 DOI: 10.1038/s41467-024-48355-5] [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: 09/13/2023] [Accepted: 04/26/2024] [Indexed: 05/30/2024] Open
Abstract
The gut microbiome (GM) modulates body weight/composition and gastrointestinal functioning; therefore, approaches targeting resident gut microbes have attracted considerable interest. Intermittent fasting (IF) and protein pacing (P) regimens are effective in facilitating weight loss (WL) and enhancing body composition. However, the interrelationships between IF- and P-induced WL and the GM are unknown. The current randomized controlled study describes distinct fecal microbial and plasma metabolomic signatures between combined IF-P (n = 21) versus a heart-healthy, calorie-restricted (CR, n = 20) diet matched for overall energy intake in free-living human participants (women = 27; men = 14) with overweight/obesity for 8 weeks. Gut symptomatology improves and abundance of Christensenellaceae microbes and circulating cytokines and amino acid metabolites favoring fat oxidation increase with IF-P (p < 0.05), whereas metabolites associated with a longevity-related metabolic pathway increase with CR (p < 0.05). Differences indicate GM and metabolomic factors play a role in WL maintenance and body composition. This novel work provides insight into the GM and metabolomic profile of participants following an IF-P or CR diet and highlights important differences in microbial assembly associated with WL and body composition responsiveness. These data may inform future GM-focused precision nutrition recommendations using larger sample sizes of longer duration. Trial registration, March 6, 2020 (ClinicalTrials.gov as NCT04327141), based on a previous randomized intervention trial.
Collapse
Affiliation(s)
- Alex E Mohr
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- Biodesign Institute Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - Karen L Sweazea
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- Biodesign Institute Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
- Center for Evolution and Medicine, College of Liberal Arts and Sciences, Arizona State University, Tempe, AZ, USA
| | - Devin A Bowes
- Biodesign Institute Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - Paniz Jasbi
- School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
- Systems Precision Engineering and Advanced Research (SPEAR), Theriome Inc., Phoenix, AZ, USA
| | - Corrie M Whisner
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- Biodesign Institute Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - Dorothy D Sears
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Rosa Krajmalnik-Brown
- Biodesign Institute Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - Yan Jin
- Center of Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Haiwei Gu
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- Center of Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Judith Klein-Seetharaman
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
| | - Karen M Arciero
- Human Nutrition and Metabolism Laboratory, Department of Health and Human Physiological Sciences, Skidmore College, Saratoga Springs, NY, USA
| | | | - Paul J Arciero
- Human Nutrition and Metabolism Laboratory, Department of Health and Human Physiological Sciences, Skidmore College, Saratoga Springs, NY, USA.
- School of Health and Rehabilitation Sciences, Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
8
|
Bin Hafeez A, Pełka K, Worobo R, Szweda P. In Silico Safety Assessment of Bacillus Isolated from Polish Bee Pollen and Bee Bread as Novel Probiotic Candidates. Int J Mol Sci 2024; 25:666. [PMID: 38203838 PMCID: PMC10780176 DOI: 10.3390/ijms25010666] [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: 12/11/2023] [Revised: 12/23/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Bacillus species isolated from Polish bee pollen (BP) and bee bread (BB) were characterized for in silico probiotic and safety attributes. A probiogenomics approach was used, and in-depth genomic analysis was performed using a wide array of bioinformatics tools to investigate the presence of virulence and antibiotic resistance properties, mobile genetic elements, and secondary metabolites. Functional annotation and Carbohydrate-Active enZYmes (CAZYme) profiling revealed the presence of genes and a repertoire of probiotics properties promoting enzymes. The isolates BB10.1, BP20.15 (isolated from bee bread), and PY2.3 (isolated from bee pollen) genome mining revealed the presence of several genes encoding acid, heat, cold, and other stress tolerance mechanisms, adhesion proteins required to survive and colonize harsh gastrointestinal environments, enzymes involved in the metabolism of dietary molecules, antioxidant activity, and genes associated with the synthesis of vitamins. In addition, genes responsible for the production of biogenic amines (BAs) and D-/L-lactate, hemolytic activity, and other toxic compounds were also analyzed. Pan-genome analyses were performed with 180 Bacillus subtilis and 204 Bacillus velezensis genomes to mine for any novel genes present in the genomes of our isolates. Moreover, all three isolates also consisted of gene clusters encoding secondary metabolites.
Collapse
Affiliation(s)
- Ahmer Bin Hafeez
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Ul. G. Narutowicza 11/12, 80-233 Gdańsk, Poland; (A.B.H.); (K.P.)
| | - Karolina Pełka
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Ul. G. Narutowicza 11/12, 80-233 Gdańsk, Poland; (A.B.H.); (K.P.)
| | - Randy Worobo
- Department of Food Science, Cornell University, Ithaca, NY 14853, USA;
| | - Piotr Szweda
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdańsk University of Technology, Ul. G. Narutowicza 11/12, 80-233 Gdańsk, Poland; (A.B.H.); (K.P.)
| |
Collapse
|
9
|
Liang X, Zhai Z, Ren F, Jie Y, Kim SK, Niu KM, Wu X. Metagenomic characterization of the cecal microbiota community and functions in finishing pigs fed fermented Boehmeria nivea. Front Vet Sci 2023; 10:1253778. [PMID: 37841475 PMCID: PMC10569026 DOI: 10.3389/fvets.2023.1253778] [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: 07/06/2023] [Accepted: 09/05/2023] [Indexed: 10/17/2023] Open
Abstract
Ramie (Boehmeria nivea, BN) is used as livestock forage through suitable silage fermentation owing to its nutritional value. To date, relatively few studies have investigated the effects of dietary fermented BN (FBN) on gut health in finishing pigs. The aim of the present study was to investigate the effects of dietary supplementation with 20% FBN on intestinal morphology, gene expression, and the functional response of the gut microbiota in finishing pigs. We found that FBN did not significantly affect serum antioxidant enzyme activities, ileal morphology, or the expression of genes encoding antioxidant enzymes, inflammatory cytokines, or tight junction proteins in the liver of the pigs. However, the gene expression levels of aryl hydrocarbon receptor (AHR) and interleukin 6 (IL6) were significantly downregulated in the ileum. A metagenomic analysis demonstrated that, compared with that seen in the control group, the cecal microbiota of pigs in the FBN treatment group was more closely clustered and contained a greater number of unique microbes. Bacteria were the predominant kingdom in the cecal microbiota, while Firmicutes, Bacteroidetes, and Proteobacteria were the dominant phyla, and Streptococcus, Lactobacillus, and Prevotella were the dominant genera. Dietary FBN significantly increased the abundance of the probiotic bacterium Roseburia inulinivorans (p < 0.05). Functional analysis of the cecal microbiota showed that ABC transporter levels and glycolysis/gluconeogenesis-associated functions were diminished in FBN-fed pigs. Meanwhile, CAZyme analysis revealed that dietary FBN significantly downregulated the contents of carbohydrate-active enzymes, such as GT2, GH1, GH25, and GH13_31. In addition, cytochrome P450 analysis revealed that the abundance of CYP51 and CYP512 decreased with FBN treatment. An assessment of antibiotic resistance based on the Comprehensive Antibiotic Resistance Database (CARD) annotation indicated that the cecal microbes from pigs in the FBN treatment group had increased resistance to lincosamide, streptogramin, and chloramphenicol and reduced resistance to amikacin, isepamicin, neomycin, lividomycin, gentamicin, paromomycin, ribostamycin, and butirosin. Finally, virulence factor-related analysis showed that putative hemolysin-associated functions were decreased, whereas fibronectin-binding protein, flagella, and alginate-associated functions were increased. Taken together, our data showed that FBN supplementation exerted only minor effects on intestinal morphology and microbial community composition, suggesting that it is potentially safe for use as a supplement in the diets of finishing pigs. However, more studies are needed to validate its functionality.
Collapse
Affiliation(s)
- Xiaoxiao Liang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, China
| | - Zhenya Zhai
- Jiangxi Functional Feed Additive Engineering Laboratory, Institute of Biological Resource, Jiangxi Academy of Sciences, Nanchang, China
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
| | - Fengyun Ren
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yucheng Jie
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Soo-Ki Kim
- Department of Animal Science and Technology, Konkuk University, Seoul, Republic of Korea
| | - Kai-Min Niu
- Jiangxi Functional Feed Additive Engineering Laboratory, Institute of Biological Resource, Jiangxi Academy of Sciences, Nanchang, China
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
| | - Xin Wu
- Jiangxi Functional Feed Additive Engineering Laboratory, Institute of Biological Resource, Jiangxi Academy of Sciences, Nanchang, China
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| |
Collapse
|
10
|
O’Reilly C, Mills S, Rea MC, Lavelle A, Ghosh S, Hill C, Ross RP. Interplay between inflammatory bowel disease therapeutics and the gut microbiome reveals opportunities for novel treatment approaches. MICROBIOME RESEARCH REPORTS 2023; 2:35. [PMID: 37849974 PMCID: PMC7615213 DOI: 10.20517/mrr.2023.41] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/19/2023]
Abstract
Inflammatory bowel disease (IBD) is a complex heterogeneous disorder defined by recurring chronic inflammation of the gastrointestinal tract, attributed to a combination of factors including genetic susceptibility, altered immune response, a shift in microbial composition/microbial insults (infection/exposure), and environmental influences. Therapeutics generally used to treat IBD mainly focus on the immune response and include non-specific anti-inflammatory and immunosuppressive therapeutics and targeted therapeutics aimed at specific components of the immune system. Other therapies include exclusive enteral nutrition and emerging stem cell therapies. However, in recent years, scientists have begun to examine the interplay between these therapeutics and the gut microbiome, and we present this information here. Many of these therapeutics are associated with alterations to gut microbiome composition and functionality, often driving it toward a "healthier profile" and preclinical studies have revealed that such alterations can play an important role in therapeutic efficacy. The gut microbiome can also improve or hinder IBD therapeutic efficacy or generate undesirable metabolites. For certain IBD therapeutics, the microbiome composition, particularly before treatment, may serve as a biomarker of therapeutic efficacy. Utilising this information and manipulating the interactions between the gut microbiome and IBD therapeutics may enhance treatment outcomes in the future and bring about new opportunities for personalised, precision medicine.
Collapse
Affiliation(s)
- Catherine O’Reilly
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61C996, Ireland
- Microbiology Department, University College Cork, Co. Cork T12TP07, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
- Authors contributed equally
| | - Susan Mills
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
- Authors contributed equally
| | - Mary C. Rea
- Food Biosciences Department, Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61C996, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| | - Aonghus Lavelle
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| | - Subrata Ghosh
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| | - Colin Hill
- Microbiology Department, University College Cork, Co. Cork T12TP07, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| | - R. Paul Ross
- Microbiology Department, University College Cork, Co. Cork T12TP07, Ireland
- APC Microbiome Ireland, University College Cork, Co. Cork T12YT20, Ireland
| |
Collapse
|
11
|
Takahashi H, Fujii T, Yamakawa S, Yamada C, Fujiki K, Kondo N, Funasaka K, Hirooka Y, Tochio T. Combined oral intake of short and long fructans alters the gut microbiota in food allergy model mice and contributes to food allergy prevention. BMC Microbiol 2023; 23:266. [PMID: 37737162 PMCID: PMC10515425 DOI: 10.1186/s12866-023-03021-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/14/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND It has become clear that the intestinal microbiota plays a role in food allergies. The objective of this study was to assess the food allergy-preventive effects of combined intake of a short fructan (1-kestose [Kes]) and a long fructan (inulin ([Inu]) in an ovalbumin (OVA)-induced food allergy mouse model. RESULTS Oral administration of fructans lowered the allergenic symptom score and alleviated the decreases in rectal temperature and total IgA levels and increases in OVA-specific IgE and IgA levels induced by high-dose OVA challenge, and in particular, combined intake of Kes and Inu significantly suppressed the changes in all these parameters. The expression of the pro-inflammatory cytokine IL-4, which was increased in the allergy model group, was significantly suppressed by fructan administration, and the expression of the anti-inflammatory cytokine IL-10 was significantly increased upon Kes administration. 16 S rRNA amplicon sequencing of the gut microbiota and beta diversity analysis revealed that fructan administration may induce gut microbiota resistance to food allergy sensitization, rather than returning the gut microbiota to a non-sensitized state. The relative abundances of the genera Parabacteroides B 862,066 and Alloprevotella, which were significantly reduced by food allergy sensitization, were restored by fructan administration. In Parabacteroides, the relative abundances of Parabacteroides distasonis, Parabacteroides goldsteinii, and their fructan-degrading glycoside hydrolase family 32 gene copy numbers were increased upon Kes or Inu administration. The concentrations of short-chain fatty acids (acetate and propionate) and lactate were increased by fructan administration, especially significantly in the Kes + Inu, Kes, and Inu-fed (Inu, Kes + Inu) groups. CONCLUSION Combined intake of Kes and Inu suppressed allergy scores more effectively than single intake, suggesting that Kes and Inu have different allergy-preventive mechanisms. This indicates that the combined intake of these short and long fructans may have an allergy-preventive benefit.
Collapse
Affiliation(s)
- Hideaki Takahashi
- Graduate School of Nutritional Sciences, Nagoya University of Arts and Sciences, Nisshin, Aichi, Japan
| | - Tadashi Fujii
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan.
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake, Aichi, Japan.
| | - Saki Yamakawa
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
- Research and Development Division, Itochu Sugar Co., Ltd., Hekinan, Aichi, Japan
- WELLNEO SUGAR Co., Ltd., Tokyo, Japan
| | - Chikako Yamada
- Graduate School of Nutritional Sciences, Nagoya University of Arts and Sciences, Nisshin, Aichi, Japan
| | - Kotoyo Fujiki
- Graduate School of Nutritional Sciences, Nagoya University of Arts and Sciences, Nisshin, Aichi, Japan
| | - Nobuhiro Kondo
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
- Research and Development Division, Itochu Sugar Co., Ltd., Hekinan, Aichi, Japan
- WELLNEO SUGAR Co., Ltd., Tokyo, Japan
| | - Kohei Funasaka
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
| | - Yoshiki Hirooka
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake, Aichi, Japan
| | - Takumi Tochio
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
12
|
de Souza Vilela J, Kheravii SK, Sharma Bajagai Y, Kolakshyapati M, Zimazile Sibanda T, Wu SB, Andrew NR, Ruhnke I. Inclusion of up to 20% Black Soldier Fly larvae meal in broiler chicken diet has a minor effect on caecal microbiota. PeerJ 2023; 11:e15857. [PMID: 37744229 PMCID: PMC10516104 DOI: 10.7717/peerj.15857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/16/2023] [Indexed: 09/26/2023] Open
Abstract
Background The Black Soldier Fly larvae (BSFL) are a source of nutrients and bioactive compounds in broiler diets. Some components of the BSFL may serve as a prebiotic or may impact the intestinal microbiota of the broilers by other modes of action, which in turn can affect the health and performance of broilers. Here, we investigate the impact of up to 20% BSFL in broiler diets on the diversity and composition of the broiler's microbiota. Methods Four hundred broilers were fed five iso-nutritious experimental diets with increasing levels of BSFL meal reaching 0%, 5%, 10%, 15%, 20% in the finisher diets. Eight caecal content samples coming from each of the eight replicates per treatment were collected at two time points (day 21 and day 42) for DNA extraction and sequencing of the V3-V4 regions using Illumina MiSeq 2 × 300 bp pair-end sequencing with 341f and 805r primers. Analysis of variance and Spearman's correlation were performed, while QIIME2, DADA2, and Calypso were used for data analysis. Results When broilers were 21 days of age, the abundance of two groups of sequence variants representing Enterococcus and unclassified Christensenellaceae was significantly lower (p-value = 0.048 and p-value = 0.025, respectively) in the 20% BSFL group compared to the 0% BSFL group. There was no relevant alteration in the microbiota diversity at that stage. On day 42, the Spearman correlation analysis demonstrated that the sequence variants representing the genus Coprococcus showed a negative relationship with the BSFL inclusion levels (p-value = 0.043). The sequence variants representing the genus Roseburia and Dehalobacterium demonstrated a positive relationship with the BSFL dietary inclusion (p-value = 0.0069 and p-value = 0.0034, respectively). There was a reduction in the dissimilarity index (ANOSIM) caused by the 20% BSFL dietary inclusion. Conclusion The addition of up to 20% BSFL in broiler diets did not affect the overall caeca microbiota diversity or composition at day 21. On day 42, there was a reduction in the beta diversity caused by the 20% BSFL dietary inclusion. The abundance of the bacterial group Roseburia was increased by the BSFL dietary inclusion, and it may be beneficial to broiler immunity and performance.
Collapse
Affiliation(s)
- Jessica de Souza Vilela
- School of Environmental and Rural Science, University of New England, Armidale, NSW, Australia
| | - Sarbast K. Kheravii
- School of Environmental and Rural Science, University of New England, Armidale, NSW, Australia
| | - Yadav Sharma Bajagai
- Institute for Future Farming Systems, Central Queensland University, Rockhampton, Queensland, Australia
| | - Manisha Kolakshyapati
- School of Environmental and Rural Science, University of New England, Armidale, NSW, Australia
| | | | - Shu-Biao Wu
- School of Environmental and Rural Science, University of New England, Armidale, NSW, Australia
| | - Nigel R. Andrew
- School of Environmental and Rural Science, University of New England, Armidale, NSW, Australia
- Faculty of Science and Engineering, Southern Cross University, Lismore, NSW, Australia
| | - Isabelle Ruhnke
- School of Environmental and Rural Science, University of New England, Armidale, NSW, Australia
| |
Collapse
|
13
|
Wang Z, Guo M, Li J, Jiang C, Yang S, Zheng S, Li M, Ai X, Xu X, Zhang W, He X, Wang Y, Chen Y. Composition and functional profiles of gut microbiota reflect the treatment stage, severity, and etiology of acute pancreatitis. Microbiol Spectr 2023; 11:e0082923. [PMID: 37698429 PMCID: PMC10580821 DOI: 10.1128/spectrum.00829-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 07/13/2023] [Indexed: 09/13/2023] Open
Abstract
Acute pancreatitis (AP) is a type of digestive system disease with high mortality. Previous studies have shown that gut microbiota can participate in developing and treating acute pancreatitis by affecting the host's metabolism. In this study, we followed 20 AP patients to generate longitudinal gut microbiota profiles and activity during disease (before treatment, on the third day of treatment, and 1 month after discharge). We analyzed species composition and metabolic pathways' changes across the treatment phase, severity, and etiology. The diversity of the gut microbiome of patients with AP did not show much variation with treatment. In contrast, the metabolic functions of the gut microbiota, such as the essential chemical reactions that produce energy and maintain life, were partially reinstated after treatment. The severe AP (SAP) patients contained less beneficial bacteria (i.e., Bacteroides xylanisolvens, Clostridium lavalense, and Roseburia inulinivorans) and weaker sugar degradation function than mild AP patients before treatment. Moreover, etiology was one of the drivers of gut microbiome composition and explained the 3.54% variation in species' relative abundance. The relative abundance of pathways related to lipid synthesis was higher in the gut of hyperlipidemia AP patients than in biliary AP patients. The composition and functional profiles of the gut microbiota reflect the severity and etiology of AP. Otherwise, we also identified bacterial species associated with SAP, i.e., Oscillibacter sp. 57_20, Parabacteroides johnsonii, Bacteroides stercoris, Methanobrevibacter smithii, Ruminococcus lactaris, Coprococcus comes, and Dorea formicigenerans, which have the potential to identify the SAP at an early stage. IMPORTANCE Acute pancreatitis (AP) is a type of digestive system disease with high mortality. Previous studies have shown that gut microbiota can participate in the development and treatment of acute pancreatitis by affecting the host's metabolism. However, fewer studies acquired metagenomic sequencing data to associate species to functions intuitively and performed longitudinal analysis to explore how gut microbiota influences the development of AP. We followed 20 AP patients to generate longitudinal gut microbiota profiles and activity during disease and studied the differences in intestinal flora under different severities and etiologies. We have two findings. First, the gut microbiota profile has the potential to identify the severity and etiology of AP at an early stage. Second, gut microbiota likely acts synergistically in the development of AP. This study provides a reference for characterizing the driver flora of severe AP to identify the severity of acute pancreatitis at an early stage.
Collapse
Affiliation(s)
- Zhenjiang Wang
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Mingyi Guo
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Jing Li
- School of Management, University of Science and Technology of China, Hefei, Anhui, China
- Department of Research and Development, Shenzhen Byoryn Technology Co., Ltd., Shenzhen, China
| | - Chuangming Jiang
- Department of Gastroenterology, Gaolangang Branch of Zhuhai People’s Hospital (Hospital of Gaolangang), Zhuhai, China
| | - Sen Yang
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Shizhuo Zheng
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Mingzhe Li
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Xinbo Ai
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Xiaohong Xu
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| | - Wenbo Zhang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Xingxiang He
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yinan Wang
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yuping Chen
- Department of Gastroenterology, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, China
| |
Collapse
|
14
|
Bester A, O'Brien M, Cotter PD, Dam S, Civai C. Shotgun Metagenomic Sequencing Revealed the Prebiotic Potential of a Fruit Juice Drink with Fermentable Fibres in Healthy Humans. Foods 2023; 12:2480. [PMID: 37444219 DOI: 10.3390/foods12132480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/13/2023] [Accepted: 06/08/2023] [Indexed: 07/15/2023] Open
Abstract
Fibre-based dietary interventions are at the forefront of gut microbiome modulation research, with a wealth of 16S rRNA information to demonstrate the prebiotic effects of isolated fibres. However, there is a distinct lack of data relating to the effect of a combination of soluble and insoluble fibres in a convenient-to-consume fruit juice food matrix on gut microbiota structure, diversity, and function. Here, we aimed to determine the impact of the MOJU Prebiotic Shot, an apple, lemon, ginger, and raspberry fruit juice drink blend containing chicory inulin, baobab, golden kiwi, and green banana powders, on gut microbiota structure and function. Healthy adults (n = 20) were included in a randomised, double-blind, placebo-controlled, cross-over study, receiving 60 mL MOJU Prebiotic Shot or placebo (without the fibre mix) for 3 weeks with a 3-week washout period between interventions. Shotgun metagenomics revealed significant between-group differences in alpha and beta diversity. In addition, the relative abundance of the phyla Actinobacteria and Desulfobacteria was significantly increased as a result of the prebiotic intervention. Nine species were observed to be differentially abundant (uncorrected p-value of <0.05) as a result of the prebiotic treatment. Of these, Bifidobacterium adolescentis and CAG-81 sp900066785 (Lachnospiraceae) were present at increased abundance relative to baseline. Additionally, KEGG analysis showed an increased abundance in pathways associated with arginine biosynthesis and phenylacetate degradation during the prebiotic treatment. Our results show the effects of the daily consumption of 60 mL MOJU Prebiotic Shot for 3 weeks and provide insight into the functional potential of B. adolescentis.
Collapse
Affiliation(s)
- Adri Bester
- London Agri Food Innovation Clinic (LAFIC), School of Applied Sciences, London South Bank University, London SE1 0AA, UK
| | | | | | | | - Claudia Civai
- London Agri Food Innovation Clinic (LAFIC), School of Applied Sciences, London South Bank University, London SE1 0AA, UK
| |
Collapse
|
15
|
Singh RP, Niharika J, Thakur R, Wagstaff BA, Kumar G, Kurata R, Patel D, Levy CW, Miyazaki T, Field RA. Utilization of dietary mixed-linkage β-glucans by the Firmicute Blautia producta. J Biol Chem 2023; 299:104806. [PMID: 37172725 PMCID: PMC10318527 DOI: 10.1016/j.jbc.2023.104806] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
The β-glucans are structurally varied, naturally occurring components of the cell walls, and storage materials of a variety of plant and microbial species. In the human diet, mixed-linkage glucans [MLG - β-(1,3/4)-glucans] influence the gut microbiome and the host immune system. Although consumed daily, the molecular mechanism by which human gut Gram-positive bacteria utilize MLG largely remains unknown. In this study, we used Blautia producta ATCC 27340 as a model organism to develop an understanding of MLG utilization. B. producta encodes a gene locus comprising a multi-modular cell-anchored endo-glucanase (BpGH16MLG), an ABC transporter, and a glycoside phosphorylase (BpGH94MLG) for utilizing MLG, as evidenced by the upregulation of expression of the enzyme- and solute binding protein (SBP)-encoding genes in this cluster when the organism is grown on MLG. We determined that recombinant BpGH16MLG cleaved various types of β-glucan, generating oligosaccharides suitable for cellular uptake by B. producta. Cytoplasmic digestion of these oligosaccharides is then performed by recombinant BpGH94MLG and β-glucosidases (BpGH3-AR8MLG and BpGH3-X62MLG). Using targeted deletion, we demonstrated BpSBPMLG is essential for B. producta growth on barley β-glucan. Furthermore, we revealed that beneficial bacteria, such as Roseburia faecis JCM 17581T, Bifidobacterium pseudocatenulatum JCM 1200T, Bifidobacterium adolescentis JCM 1275T, and Bifidobacterium bifidum JCM 1254, can also utilize oligosaccharides resulting from the action of BpGH16MLG. Disentangling the β-glucan utilizing the capability of B. producta provides a rational basis on which to consider the probiotic potential of this class of organism.
Collapse
Affiliation(s)
- Ravindra Pal Singh
- Department of Industrial Biotechnology, Gujarat Biotechnology University, Near Gujarat International Finance Tec (GIFT)-City, Gandhinagar, Gujarat, India; Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, SAS Nagar, Punjab, India.
| | - Jayashree Niharika
- Department of Industrial Biotechnology, Gujarat Biotechnology University, Near Gujarat International Finance Tec (GIFT)-City, Gandhinagar, Gujarat, India; Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, SAS Nagar, Punjab, India
| | - Raksha Thakur
- Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, SAS Nagar, Punjab, India
| | - Ben A Wagstaff
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Gulshan Kumar
- Division of Food and Nutritional Biotechnology, National Agri-Food Biotechnology Institute, SAS Nagar, Punjab, India
| | - Rikuya Kurata
- Department of Agriculture, Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka City, Shizuoka, Japan
| | - Dhaval Patel
- Department of Industrial Biotechnology, Gujarat Biotechnology University, Near Gujarat International Finance Tec (GIFT)-City, Gandhinagar, Gujarat, India
| | - Colin W Levy
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Takatsugu Miyazaki
- Department of Agriculture, Graduate School of Integrated Science and Technology, Shizuoka University, Shizuoka City, Shizuoka, Japan; Research Institute of Green Science and Technology, Shizuoka University, Shizuoka City, Shizuoka, Japan
| | - Robert A Field
- Department of Chemistry and Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK.
| |
Collapse
|
16
|
Slevin E, Koyama S, Harrison K, Wan Y, Klaunig JE, Wu C, Shetty AK, Meng F. Dysbiosis in gastrointestinal pathophysiology: Role of the gut microbiome in Gulf War Illness. J Cell Mol Med 2023; 27:891-905. [PMID: 36716094 PMCID: PMC10064030 DOI: 10.1111/jcmm.17631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 01/31/2023] Open
Abstract
Gulf War Illness (GWI) has been reported in 25%-35% of veterans returned from the Gulf war. Symptoms of GWI are varied and include both neurological and gastrointestinal symptoms as well as chronic fatigue. Development of GWI has been associated with chemical exposure particularly with exposure to pyridostigmine bromide (PB) and permethrin. Recent studies have found that the pathology of GWI is connected to changes in the gut microbiota, that is the gut dysbiosis. In studies using animal models, the exposure to PB and permethrin resulted in similar changes in the gut microbiome as these found in GW veterans with GWI. Studies using animal models have also shown that phytochemicals like curcumin are beneficial in reducing the symptoms and that the extracellular vesicles (EV) released from gut bacteria and from the intestinal epithelium can both promote diseases and suppress diseases through the intercellular communication mechanisms. The intestinal epithelium cells produce EVs and these EVs of intestinal epithelium origin are found to suppress inflammatory bowel disease severity, suggesting the benefits of utilizing EV in treatments. On the contrary, EV from the plasma of septic mice enhanced the level of proinflammatory cytokines in vitro and neutrophils and macrophages in vivo, suggesting differences in the EV depending on the types of cells they were originated and/or influences of environmental changes. These studies suggest that targeting the EV that specifically have positive influences may become a new therapeutic strategy in the treatment of veterans with GWI.
Collapse
Affiliation(s)
- Elise Slevin
- Division of Gastroenterology and Hepatology, Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
- Richard L. Roudebush VA Medical CenterIndianapolisIndianaUSA
| | - Sachiko Koyama
- Division of Gastroenterology and Hepatology, Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
- Richard L. Roudebush VA Medical CenterIndianapolisIndianaUSA
| | - Kelly Harrison
- Department of Transplant SurgeryBaylor Scott & White Memorial HospitalTempleTexasUSA
| | - Ying Wan
- Department of Pathophysiology, School of Basic Medical ScienceSouthwest Medical UniversityLuzhouChina
| | - James E. Klaunig
- Laboratory of Investigative Toxicology and Pathology, Department of Environmental and Occupational Health, Indiana School of Public HealthIndiana UniversityBloomingtonIndianaUSA
| | - Chaodong Wu
- Department of NutritionTexas A&M UniversityCollege StationTexasUSA
| | - Ashok K. Shetty
- Department of Molecular and Cellular MedicineInstitute for Regenerative Medicine, Texas A&M College of MedicineCollege StationTexasUSA
| | - Fanyin Meng
- Division of Gastroenterology and Hepatology, Department of MedicineIndiana University School of MedicineIndianapolisIndianaUSA
- Richard L. Roudebush VA Medical CenterIndianapolisIndianaUSA
| |
Collapse
|
17
|
Donovan M, Mackey CS, Lynch MDJ, Platt GN, Brown AN, Washburn BK, Trickey DJ, Curtis JT, Liu Y, Charles TC, Wang Z, Jones KM. Limosilactobacillus reuteri administration alters the gut-brain-behavior axis in a sex-dependent manner in socially monogamous prairie voles. Front Microbiol 2023; 14:1015666. [PMID: 36846764 PMCID: PMC9945313 DOI: 10.3389/fmicb.2023.1015666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 01/05/2023] [Indexed: 02/11/2023] Open
Abstract
Research on the role of gut microbiota in behavior has grown dramatically. The probiotic L. reuteri can alter social and stress-related behaviors - yet, the underlying mechanisms remain largely unknown. Although traditional laboratory rodents provide a foundation for examining the role of L. reuteri on the gut-brain axis, they do not naturally display a wide variety of social behaviors. Using the highly-social, monogamous prairie vole (Microtus ochrogaster), we examined the effects of L. reuteri administration on behaviors, neurochemical marker expression, and gut-microbiome composition. Females, but not males, treated with live L. reuteri displayed lower levels of social affiliation compared to those treated with heat-killed L. reuteri. Overall, females displayed a lower level of anxiety-like behaviors than males. Live L. reuteri-treated females had lower expression of corticotrophin releasing factor (CRF) and CRF type-2-receptor in the nucleus accumbens, and lower vasopressin 1a-receptor in the paraventricular nucleus of the hypothalamus (PVN), but increased CRF in the PVN. There were both baseline sex differences and sex-by-treatment differences in gut microbiome composition. Live L. reuteri increased the abundance of several taxa, including Enterobacteriaceae, Lachnospiraceae NK4A136, and Treponema. Interestingly, heat-killed L. reuteri increased abundance of the beneficial taxa Bifidobacteriaceae and Blautia. There were significant correlations between changes in microbiota, brain neurochemical markers, and behaviors. Our data indicate that L. reuteri impacts gut microbiota, gut-brain axis and behaviors in a sex-specific manner in socially-monogamous prairie voles. This demonstrates the utility of the prairie vole model for further examining causal impacts of microbiome on brain and behavior.
Collapse
Affiliation(s)
- Meghan Donovan
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, United States
- Rocky Mountain Mental Illness Research Education and Clinical Center, Rocky Mountain Regional VA Medical Center, Aurora, CO, United States
- Department of Physical Medicine and Rehabilitation, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Calvin S. Mackey
- Department of Biological Science, Florida State University, Tallahassee, FL, United States
| | - Michael D. J. Lynch
- Metagenom Bio Life Science Inc, Waterloo, ON, Canada
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Grayson N. Platt
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, United States
- Department of Biological Science, Florida State University, Tallahassee, FL, United States
| | - Amber N. Brown
- Department of Biological Science Core Facilities, Florida State University, Tallahassee, FL, United States
| | - Brian K. Washburn
- Department of Biological Science Core Facilities, Florida State University, Tallahassee, FL, United States
| | - Darryl J. Trickey
- Department of Biological Science, Florida State University, Tallahassee, FL, United States
| | - J. Thomas Curtis
- Department of Pharmacology and Physiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Yan Liu
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Trevor C. Charles
- Metagenom Bio Life Science Inc, Waterloo, ON, Canada
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, United States
| | - Kathryn M. Jones
- Department of Biological Science, Florida State University, Tallahassee, FL, United States
| |
Collapse
|
18
|
Meng X, Zheng J, Wang F, Zheng J, Yang D. Dietary fiber chemical structure determined gut microbiota dynamics. IMETA 2022; 1:e64. [PMID: 38867894 PMCID: PMC10989905 DOI: 10.1002/imt2.64] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/13/2022] [Accepted: 11/06/2022] [Indexed: 06/14/2024]
Abstract
Precision modulation of gut microbiota requires elucidation of the relation between dietary fiber intake and gut microbe dynamics. However, current studies on this aspect are few due to many technical limitations. Here, we used Caenorhabditis elegans to minimize the complicated host-microbial factors and to find the relation between dietary fiber chemical structures and gut microbiota dynamics. The Allium schoenoprasum polysaccharide (AssP) structure was elucidated and used as the complex dietary fiber against the simple fiber inulin. In vitro bacterial growth and genome analysis indicated that AssP supports bacterial growth better than inulin, while in vivo gut microbiota analysis of C. elegans fed with AssP showed that microbiota richness increased significantly compared with those fed with inulin. It is concluded that the more complex the dietary fiber chemical structure, the more gut bacteria growth it supports. Together with the community bacterial interactions that alter their abundances in vivo, these factors regulate gut microbiota synergistically.
Collapse
Affiliation(s)
- Xin Meng
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science & Nutritional EngineeringChina Agricultural UniversityBeijingChina
| | - Jun Zheng
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science & Nutritional EngineeringChina Agricultural UniversityBeijingChina
| | - Fengqiao Wang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science & Nutritional EngineeringChina Agricultural UniversityBeijingChina
| | - Jie Zheng
- Center for Food Safety and Applied NutritionU.S. Food and Drug AdministrationCollege ParkMarylandUSA
| | - Dong Yang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science & Nutritional EngineeringChina Agricultural UniversityBeijingChina
| |
Collapse
|
19
|
Pezzuto JM, Dave A, Park EJ, Beyoğlu D, Idle JR. Short-Term Grape Consumption Diminishes UV-Induced Skin Erythema. Antioxidants (Basel) 2022; 11:2372. [PMID: 36552580 PMCID: PMC9774720 DOI: 10.3390/antiox11122372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/17/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Over three million Americans are affected by skin cancer each year, largely as a result of exposure to sunlight. The purpose of this study was to determine the potential of grape consumption to modulate UV-induced skin erythema. With 29 human volunteers, we report that nine demonstrated greater resistance to UV irradiation of the skin after consuming the equivalent of three servings of grapes per day for two weeks. We further explored any potential relationship to the gut-skin axis. Alpha- and beta-diversity of the gut microbiome were not altered, but grape consumption modulated microbiota abundance, enzyme levels, and KEGG pathways. Striking differences in the microbiome and metabolome were discerned when comparing the nine individuals showing greater UV resistance with the 20 non-responders. Notably, three urinary metabolites, 2'-deoxyribonic acid, 3-hydroxyphenyl acetic and scyllo-inositol, were depressed in the UV-resistant group. A ROC curve revealed a 71.8% probability that measurement of urinary 2'-deoxyribonic acid identifies a UV skin non-responder. 2'-Deoxyribonic acid is cleaved from the DNA backbone by reactive oxygen species. Three of the nine subjects acquiring UV resistance following grape consumption showed a durable response, and these three demonstrated unique microbiomic and metabolomic profiles. Variable UV skin sensitivity was likely due to glutathione S-transferase polymorphisms. We conclude that a segment of the population is capable of demonstrating greater resistance to a dermal response elicited by UV irradiation as a result of grape consumption. It is uncertain if modulation of the gut-skin axis leads to enhanced UV resistance, but there is correlation. More broadly, it is reasonable to expect that these mechanisms relate to other health outcomes anticipated to result from grape consumption.
Collapse
Affiliation(s)
- John M. Pezzuto
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
- Department of Medicine, UMass Chan Medical School—Baystate, Springfield, MA 01199, USA
| | - Asim Dave
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Eun-Jung Park
- Division of Pharmaceutical Sciences, Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY 11201, USA
| | - Diren Beyoğlu
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
| | - Jeffrey R. Idle
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
| |
Collapse
|
20
|
Butyrate-producing colonic clostridia: picky glycan utilization specialists. Essays Biochem 2022; 67:415-428. [PMID: 36350044 DOI: 10.1042/ebc20220125] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/18/2022] [Accepted: 10/21/2022] [Indexed: 11/10/2022]
Abstract
Abstract
Butyrate-producing human gut microbiota members are recognized for their strong association with a healthy immune-homeostasis and protection from inflammatory disorders and colorectal cancer. These effects are attributed to butyrate, the terminal electron sink of glycan fermentation by prevalent and abundant colonic Firmicutes from the Lachnospiraceae and Oscillospiraceae families. Remarkably, our insight into the glycan utilization mechanisms and preferences of butyrogenic Firmicutes remains very limited as compared with other gut symbionts, especially from the Bacteroides, Bifidobacterium, and Lactobacillus genera. Here, we summarize recent findings on the strategies that colonic butyrate producers have evolved to harvest energy from major dietary fibres, especially plant structural and storage glycans, such as resistant starch, xylans, and mannans. Besides dietary fibre, we also present the unexpected discovery of a conserved protein apparatus that confers the growth of butyrate producers on human milk oligosaccharides (HMOs), which are unique to mother’s milk. The dual dietary fibre/HMO utilization machinery attests the adaptation of this group to both the infant and adult guts. These finding are discussed in relation to the early colonization of butyrogenic bacteria and the maturation of the microbiota during the transition from mother’s milk to solid food. To date, the described butyrogenic Firmicutes are glycan utilization specialists that target only a few glycans in a highly competitive manner relying on co-regulated glycan utilization loci. We describe the common pillars of this machinery, highlighting butyrate producers as a source for discovery of biochemically and structurally novel carbohydrate active enzymes.
Collapse
|
21
|
Kang JW, Tang X, Walton CJ, Brown MJ, Brewer RA, Maddela RL, Zheng JJ, Agus JK, Zivkovic AM. Multi-Omic Analyses Reveal Bifidogenic Effect and Metabolomic Shifts in Healthy Human Cohort Supplemented With a Prebiotic Dietary Fiber Blend. Front Nutr 2022; 9:908534. [PMID: 35782954 PMCID: PMC9248813 DOI: 10.3389/fnut.2022.908534] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/20/2022] [Indexed: 12/14/2022] Open
Abstract
Dietary fiber, a nutrient derived mainly from whole grains, vegetables, fruits, and legumes, is known to confer a number of health benefits, yet most Americans consume less than half of the daily recommended amount. Convenience and affordability are key factors determining the ability of individuals to incorporate fiber-rich foods into their diet, and many Americans struggle to access, afford, and prepare foods rich in fiber. The objective of this clinical study was to test the changes in microbial community composition, human metabolomics, and general health markers of a convenient, easy to use prebiotic supplement in generally healthy young participants consuming a diet low in fiber. Twenty healthy adults participated in this randomized, placebo-controlled, double-blind, crossover study which was registered at clinicaltrials.gov as NCT03785860. During the study participants consumed 12 g of a prebiotic fiber supplement and 12 g of placebo daily as a powder mixed with water as part of their habitual diet in randomized order for 4 weeks, with a 4-week washout between treatment arms. Fecal microbial DNA was extracted and sequenced by shallow shotgun sequencing on an Illumina NovaSeq. Plasma metabolites were detected using liquid chromatography–mass spectrometry with untargeted analysis. The phylum Actinobacteria, genus Bifidobacterium, and several Bifidobacterium species (B. bifidum, B. adolescentis, B. breve, B. catenulatum, and B. longum) significantly increased after prebiotic supplementation when compared to the placebo. The abundance of genes associated with the utilization of the prebiotic fiber ingredients (sacA, xfp, xpk) and the production of acetate (poxB, ackA) significantly changed with prebiotic supplementation. Additionally, the abundance of genes associated with the prebiotic utilization (xfp, xpk), acetate production (ackA), and choline to betaine oxidation (gbsB) were significantly correlated with changes in the abundance of the genus Bifidobacterium in the prebiotic group. Plasma concentrations of the bacterially produced metabolite indolepropionate significantly increased. The results of this study demonstrate that an easy to consume, low dose (12 g) of a prebiotic powder taken daily increases the abundance of beneficial bifidobacteria and the production of health-promoting bacteria-derived metabolites in healthy individuals with a habitual low-fiber diet.
Collapse
Affiliation(s)
- Jea Woo Kang
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Xinyu Tang
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | | | - Mark J. Brown
- USANA Health Sciences, Inc., Salt Lake City, UT, United States
| | | | | | - Jack Jingyuan Zheng
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Joanne K. Agus
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Angela M. Zivkovic
- Department of Nutrition, University of California, Davis, Davis, CA, United States
- *Correspondence: Angela M. Zivkovic
| |
Collapse
|
22
|
Sauvaitre T, Van Herreweghen F, Delbaere K, Durif C, Van Landuyt J, Fadhlaoui K, Huille S, Chaucheyras-Durand F, Etienne-Mesmin L, Blanquet-Diot S, Van de Wiele T. Lentils and Yeast Fibers: A New Strategy to Mitigate Enterotoxigenic Escherichia coli (ETEC) Strain H10407 Virulence? Nutrients 2022; 14:nu14102146. [PMID: 35631287 PMCID: PMC9144138 DOI: 10.3390/nu14102146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 01/10/2023] Open
Abstract
Dietary fibers exhibit well-known beneficial effects on human health, but their anti-infectious properties against enteric pathogens have been poorly investigated. Enterotoxigenic Escherichia coli (ETEC) is a major food-borne pathogen that causes acute traveler’s diarrhea. Its virulence traits mainly rely on adhesion to an epithelial surface, mucus degradation, and the secretion of two enterotoxins associated with intestinal inflammation. With the increasing burden of antibiotic resistance worldwide, there is an imperious need to develop novel alternative strategies to control ETEC infections. This study aimed to investigate, using complementary in vitro approaches, the inhibitory potential of two dietary-fiber-containing products (a lentil extract and yeast cell walls) against the human ETEC reference strain H10407. We showed that the lentil extract decreased toxin production in a dose-dependent manner, reduced pro-inflammatory interleukin-8 production, and modulated mucus-related gene induction in ETEC-infected mucus-secreting intestinal cells. We also report that the yeast product reduced ETEC adhesion to mucin and Caco-2/HT29-MTX cells. Both fiber-containing products strengthened intestinal barrier function and modulated toxin-related gene expression. In a complex human gut microbial background, both products did not elicit a significant effect on ETEC colonization. These pioneering data demonstrate the promising role of dietary fibers in controlling different stages of the ETEC infection process.
Collapse
Affiliation(s)
- Thomas Sauvaitre
- UMR 454 INRAE, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France; (T.S.); (C.D.); (K.F.); (F.C.-D.); (L.E.-M.)
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium; (F.V.H.); (K.D.); (J.V.L.); (T.V.d.W.)
| | - Florence Van Herreweghen
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium; (F.V.H.); (K.D.); (J.V.L.); (T.V.d.W.)
| | - Karen Delbaere
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium; (F.V.H.); (K.D.); (J.V.L.); (T.V.d.W.)
| | - Claude Durif
- UMR 454 INRAE, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France; (T.S.); (C.D.); (K.F.); (F.C.-D.); (L.E.-M.)
| | - Josefien Van Landuyt
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium; (F.V.H.); (K.D.); (J.V.L.); (T.V.d.W.)
| | - Khaled Fadhlaoui
- UMR 454 INRAE, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France; (T.S.); (C.D.); (K.F.); (F.C.-D.); (L.E.-M.)
| | | | - Frédérique Chaucheyras-Durand
- UMR 454 INRAE, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France; (T.S.); (C.D.); (K.F.); (F.C.-D.); (L.E.-M.)
- Lallemand SAS, 19 Rue des Briquetiers, BP 59, CEDEX, F-31702 Blagnac, France
| | - Lucie Etienne-Mesmin
- UMR 454 INRAE, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France; (T.S.); (C.D.); (K.F.); (F.C.-D.); (L.E.-M.)
| | - Stéphanie Blanquet-Diot
- UMR 454 INRAE, Microbiology, Digestive Environment and Health (MEDIS), Université Clermont Auvergne, 28 Place Henri Dunant, F-63000 Clermont-Ferrand, France; (T.S.); (C.D.); (K.F.); (F.C.-D.); (L.E.-M.)
- Correspondence: ; Tel.: +33-(0)4-73-17-83-90
| | - Tom Van de Wiele
- Center for Microbial Ecology and Technology (CMET), Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium; (F.V.H.); (K.D.); (J.V.L.); (T.V.d.W.)
| |
Collapse
|
23
|
Meng Z, Ye Z, Zhu P, Zhu J, Fang S, Qiu T, Li Y, Meng L. New Developments and Opportunities of Microbiota in Treating Breast Cancers. Front Microbiol 2022; 13:818793. [PMID: 35633703 PMCID: PMC9134200 DOI: 10.3389/fmicb.2022.818793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 04/04/2022] [Indexed: 01/01/2023] Open
Abstract
Despite the prevalence of breast cancer (BC), over half of BC cases are unrelated to known risk factors, which highlights the importance of uncovering more cancer-related factors. Currently, the microbiota has been proven to be a potent modulator of the tumor environment in BC, which regulates the immune balance in tumor-related networks. Through a large amount of data accumulation, the microbiota has shown many possibilities to reveal more insights into the development or control of BC. To expand the potential benefits of patients with BC, this study discusses the distribution profile and the effect mechanism of BC-related microbiota on tumors and further discusses its impact on different tumor therapies. Finally, we summarize the possibility of targeting microbiological therapies to improve BC treatment or in combination with other therapies.
Collapse
Affiliation(s)
- Zihui Meng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Zixuan Ye
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Pengrong Zhu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | | | | | - Tianzhu Qiu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Tianzhu Qiu,
| | - Yanan Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
- Yanan Li,
| | - Lijuan Meng
- Department of Geriatric Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Lijuan Meng,
| |
Collapse
|
24
|
Park JH, Song WS, Lee J, Jo SH, Lee JS, Jeon HJ, Kwon JE, Kim YR, Baek JH, Kim MG, Yang YH, Kim BG, Kim YG. An Integrative Multiomics Approach to Characterize Prebiotic Inulin Effects on Faecalibacterium prausnitzii. Front Bioeng Biotechnol 2022; 10:825399. [PMID: 35252133 PMCID: PMC8894670 DOI: 10.3389/fbioe.2022.825399] [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] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Faecalibacterium prausnitzii, a major commensal bacterium in the human gut, is well known for its anti-inflammatory effects, which improve host intestinal health. Although several studies have reported that inulin, a well-known prebiotic, increases the abundance of F. prausnitzii in the intestine, the mechanism underlying this effect remains unclear. In this study, we applied liquid chromatography tandem mass spectrometry (LC-MS/MS)-based multiomics approaches to identify biological and enzymatic mechanisms of F. prausnitzii involved in the selective digestion of inulin. First, to determine the preference for dietary carbohydrates, we compared the growth of F. prausnitzii in several carbon sources and observed selective growth in inulin. In addition, an LC-MS/MS-based intracellular proteomic and metabolic profiling was performed to determine the quantitative changes in specific proteins and metabolites of F. prausnitzii when grown on inulin. Interestingly, proteomic analysis revealed that the putative proteins involved in inulin-type fructan utilization by F. prausnitzii, particularly β-fructosidase and amylosucrase were upregulated in the presence of inulin. To investigate the function of these proteins, we overexpressed bfrA and ams, genes encoding β-fructosidase and amylosucrase, respectively, in Escherichia coli, and observed their ability to degrade fructan. In addition, the enzyme activity assay demonstrated that intracellular fructan hydrolases degrade the inulin-type fructans taken up by fructan ATP-binding cassette transporters. Furthermore, we showed that the fructose uptake activity of F. prausnitzii was enhanced by the fructose phosphotransferase system transporter when inulin was used as a carbon source. Intracellular metabolomic analysis indicated that F. prausnitzii could use fructose, the product of inulin-type fructan degradation, as an energy source for inulin utilization. Taken together, this study provided molecular insights regarding the metabolism of F. prauznitzii for inulin, which stimulates the growth and activity of the beneficial bacterium in the intestine.
Collapse
Affiliation(s)
- Ji-Hyeon Park
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Won-Suk Song
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Jeongchan Lee
- School of Chemical and Biological Engineering, Seoul National University, Seoul, South Korea
| | - Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Jae-Seung Lee
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Hyo-Jin Jeon
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Ji-Eun Kwon
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Ye-Rim Kim
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Ji-Hyun Baek
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Min-Gyu Kim
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| | - Yung-Hun Yang
- Department of Biological Engineering, Konkuk University, Seoul, South Korea
| | - Byung-Gee Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, South Korea
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University, Seoul, South Korea
| |
Collapse
|
25
|
Tanno H, Fujii T, Hirano K, Maeno S, Tonozuka T, Sakamoto M, Ohkuma M, Tochio T, Endo A. Characterization of fructooligosaccharide metabolism and fructooligosaccharide-degrading enzymes in human commensal butyrate producers. Gut Microbes 2022; 13:1-20. [PMID: 33439065 PMCID: PMC7833758 DOI: 10.1080/19490976.2020.1869503] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [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
Butyrate produced by gut microbiota has multiple beneficial effects on host health, and oligosaccharides derived from host diets and glycans originating from host mucus are major sources of its production. A significant reduction of butyrate-producing bacteria has been reported in patients with inflammatory bowel diseases and colorectal cancers. Although gut butyrate levels are important for host health, oligosaccharide metabolic properties in butyrate producers are poorly characterized. We studied the metabolic properties of fructooligosaccharides (FOSs) and other prebiotic oligosaccharides (i.e. raffinose and xylooligosaccharides; XOSs) in gut butyrate producers. 1-Kestose (kestose) and nystose, FOSs with degrees of polymerization of 3 and 4, respectively, were also included. Fourteen species of butyrate producers were divided into four groups based on their oligosaccharide metabolic properties, which are group A (two species) metabolizing all oligosaccharides tested, group F (four species) metabolizing FOSs but not raffinose and XOSs, group XR (four species) metabolizing XOSs and/or raffinose but not FOSs, and group N (four species) metabolizing none of the oligosaccharides tested. Species assigned to groups A and XR are rich glycoside hydrolase (GH) holders, whereas those in groups F and N are the opposite. In total, 17 enzymes assigned to GH32 were observed in nine of the 14 butyrate producers tested, and species that metabolized FOSs had at least one active GH32 enzyme. The GH32 enzymes were divided into four clusters by phylogenetic analysis. Heterologous gene expression analysis revealed that the GH32 enzymes in each cluster had similar FOS degradation properties within clusters, which may be linked to the conservation/substitution of amino acids to bind with substrates in GH32 enzymes. This study provides important knowledge to understand the impact of FOS supplementation on the activation of gut butyrate producers. Abbreviations: SCFA, short chain fatty acid; FOS, fructooligosaccharide; XOS, xylooligosaccharide; CAZy, Carbohydrate Active Enzymes; CBM, carbohydrate-binding module; PUL, polysaccharide utilization locus; S6PH sucrose-6-phosphate hydrolase.
Collapse
Affiliation(s)
- Hiroki Tanno
- Department of Food, Aroma and Cosmetic Chemistry, Faculty of Bioindustry, Tokyo University of Agriculture, Hokkaido, Japan
| | | | | | - Shintaro Maeno
- Department of Food, Aroma and Cosmetic Chemistry, Faculty of Bioindustry, Tokyo University of Agriculture, Hokkaido, Japan
| | - Takashi Tonozuka
- Department of Applied Biological Science, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Mitsuo Sakamoto
- PRIME, Japan Agency for Medical Research and Development (AMED), Ibaraki, Japan,Microbe Division/Japan Collection of Microorganisms, RIKEN BioResource Research Center, Ibaraki, Japan
| | - Moriya Ohkuma
- Microbe Division/Japan Collection of Microorganisms, RIKEN BioResource Research Center, Ibaraki, Japan
| | | | - Akihito Endo
- Department of Food, Aroma and Cosmetic Chemistry, Faculty of Bioindustry, Tokyo University of Agriculture, Hokkaido, Japan,CONTACT Akihito Endo Department of Food, Aroma and Cosmetic Chemistry, Faculty of Bioindustry, Tokyo University of Agriculture, 196 Yasaka, Abashiri, Hokkaido099-2493, Japan
| |
Collapse
|
26
|
Colnet B, Sieber CMK, Perraudeau F, Leclerc M. FiberGrowth Pipeline: A Framework Toward Predicting Fiber-Specific Growth From Human Gut Bacteroidetes Genomes. Front Microbiol 2021; 12:632567. [PMID: 34690938 PMCID: PMC8527192 DOI: 10.3389/fmicb.2021.632567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 09/06/2021] [Indexed: 11/13/2022] Open
Abstract
Dietary fibers impact gut colonic health, through the production of short-chain fatty acids. A low-fiber diet has been linked to lower bacterial diversity, obesity, type 2 diabetes, and promotion of mucosal pathogens. Glycoside hydrolases (GHs) are important enzymes involved in the bacterial catabolism of fiber into short-chain fatty acids. However, the GH involved in glycan breakdown (adhesion, hydrolysis, and fermentation) are organized in polysaccharide utilization loci (PUL) with complex modularity. Our goal was to explore how the capacity of strains, from the Bacteroidetes phylum, to grow on fiber could be predicted from their genome sequences. We designed an in silico pipeline called FiberGrowth and independently validated it for seven different fibers, on 28 genomes from Bacteroidetes-type strains. To do so, we compared the existing GH annotation tools and built PUL models by using published growth and gene expression data. FiberGrowth's prediction performance in terms of true positive rate (TPR) and false positive rate (FPR) strongly depended on available data and fiber: arabinoxylan (TPR: 0.89 and FPR: 0), inulin (0.95 and 0.33), heparin (0.8 and 0.22) laminarin (0.38 and 0.17), levan (0.3 and 0.06), mucus (0.13 and 0.38), and starch (0.73 and 0.41). Being able to better predict fiber breakdown by bacterial strains would help to understand their impact on human nutrition and health. Assuming further gene expression experiment along with discoveries on structural analysis, we hope computational tools like FiberGrowth will help researchers prioritize and design in vitro experiments.
Collapse
Affiliation(s)
- Bénédicte Colnet
- Pendulum Therapeutics, San Francisco, CA, United States
- Mines Paristech, Paris, France
| | | | | | - Marion Leclerc
- Université Paris Saclay, INRAe, AgroParisTech, Micalis Institute, Jouy en Josas, France
| |
Collapse
|
27
|
Akkerman R, Logtenberg MJ, Beukema M, de Haan BJ, Faas MM, Zoetendal EG, Schols HA, de Vos P. Chicory inulin enhances fermentation of 2'-fucosyllactose by infant fecal microbiota and differentially influences immature dendritic cell and T-cell cytokine responses under normal and Th2-polarizing conditions. Food Funct 2021; 12:9018-9029. [PMID: 34382992 DOI: 10.1039/d1fo00893e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Scope: Non-digestible carbohydrates (NDCs) such as native chicory inulin and 2'-fucosyllactose (2'-FL) are added to infant formula to mimic some of the human milk oligosaccharide (HMO) functions. It is unknown whether combining inulin and 2'-FL influences their fermentation kinetics and whether the immune-modulatory effects of these NDCs are different under normal and inflammatory-prone Th2-polarizing conditions. Methods and results: We investigated the in vitro fermentation of 2'-FL and native chicory inulin, fermented individually and combined, using fecal inocula of 8-week-old infants. Native inulin was fermented in a size-dependent fashion and expedited the fermentation of 2'-FL. Fermentation of both native inulin and 2'FL increased the relative abundance of Bifidobacterium, which coincided with the production of acetate and lactate. The fermentation digesta of all fermentations differentially influenced both dendritic cell and T-cell cytokine responses under normal culture conditions or in presence of the Th2-polarizing cytokines IL-33 and TSLP, with the most pronounced effect for IL-1β in the presence of TSLP. Conclusions: Our findings show that native inulin can expedite the fermentation of 2'-FL by infant fecal microbiota and that these NDC fermentation digesta have different effects under normal and Th2-polarizing conditions, indicating that infants with different immune backgrounds might benefit from tailored NDC formulations.
Collapse
Affiliation(s)
- Renate Akkerman
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands.
| | - Madelon J Logtenberg
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands.
| | - Martin Beukema
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands.
| | - Bart J de Haan
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands.
| | - Marijke M Faas
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands.
| | - Erwin G Zoetendal
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Henk A Schols
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands.
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands.
| |
Collapse
|
28
|
Barber TM, Valsamakis G, Mastorakos G, Hanson P, Kyrou I, Randeva HS, Weickert MO. Dietary Influences on the Microbiota-Gut-Brain Axis. Int J Mol Sci 2021; 22:ijms22073502. [PMID: 33800707 PMCID: PMC8038019 DOI: 10.3390/ijms22073502] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Over unimaginable expanses of evolutionary time, our gut microbiota have co-evolved with us, creating a symbiotic relationship in which each is utterly dependent upon the other. Far from confined to the recesses of the alimentary tract, our gut microbiota engage in complex and bi-directional communication with their host, which have far-reaching implications for overall health, wellbeing and normal physiological functioning. Amongst such communication streams, the microbiota–gut–brain axis predominates. Numerous complex mechanisms involve direct effects of the microbiota, or indirect effects through the release and absorption of the metabolic by-products of the gut microbiota. Proposed mechanisms implicate mitochondrial function, the hypothalamus–pituitary–adrenal axis, and autonomic, neuro-humeral, entero-endocrine and immunomodulatory pathways. Furthermore, dietary composition influences the relative abundance of gut microbiota species. Recent human-based data reveal that dietary effects on the gut microbiota can occur rapidly, and that our gut microbiota reflect our diet at any given time, although much inter-individual variation pertains. Although most studies on the effects of dietary macronutrients on the gut microbiota report on associations with relative changes in the abundance of particular species of bacteria, in broad terms, our modern-day animal-based Westernized diets are relatively high in fats and proteins and impoverished in fibres. This creates a perfect storm within the gut in which dysbiosis promotes localized inflammation, enhanced gut wall permeability, increased production of lipopolysaccharides, chronic endotoxemia and a resultant low-grade systemic inflammatory milieu, a harbinger of metabolic dysfunction and many modern-day chronic illnesses. Research should further focus on the colony effects of the gut microbiota on health and wellbeing, and dysbiotic effects on pathogenic pathways. Finally, we should revise our view of the gut microbiota from that of a seething mass of microbes to one of organ-status, on which our health and wellbeing utterly depends. Future guidelines on lifestyle strategies for wellbeing should integrate advice on the optimal establishment and maintenance of a healthy gut microbiota through dietary and other means. Although we are what we eat, perhaps more importantly, we are what our gut microbiota thrive on and they thrive on what we eat.
Collapse
Affiliation(s)
- Thomas M. Barber
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry CV2 2DX, UK; (T.M.B.); (G.V.); (P.H.); (I.K.); (H.S.R.)
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV2 2DX, UK
| | - Georgios Valsamakis
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry CV2 2DX, UK; (T.M.B.); (G.V.); (P.H.); (I.K.); (H.S.R.)
- Endocrine Unit, 2nd Department of Obstetrics and Gynaecology and Pathology Department, Aretaieion University Hospital, Athens Medical School, 11528 Athens, Greece;
| | - George Mastorakos
- Endocrine Unit, 2nd Department of Obstetrics and Gynaecology and Pathology Department, Aretaieion University Hospital, Athens Medical School, 11528 Athens, Greece;
| | - Petra Hanson
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry CV2 2DX, UK; (T.M.B.); (G.V.); (P.H.); (I.K.); (H.S.R.)
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV2 2DX, UK
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry CV2 2DX, UK; (T.M.B.); (G.V.); (P.H.); (I.K.); (H.S.R.)
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV2 2DX, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry CV2 2DX, UK; (T.M.B.); (G.V.); (P.H.); (I.K.); (H.S.R.)
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV2 2DX, UK
- Aston Medical Research Institute, Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham B4 7ET, UK
| | - Martin O. Weickert
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism, University Hospitals Coventry and Warwickshire NHS Trust, Clifford Bridge Road, Coventry CV2 2DX, UK; (T.M.B.); (G.V.); (P.H.); (I.K.); (H.S.R.)
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV2 2DX, UK
- Centre for Sport, Exercise and Life Sciences, Faculty of Health & Life Sciences, Coventry University, Coventry CV1 5FB, UK
- Correspondence:
| |
Collapse
|
29
|
Jiang H, Fang S, Yang H, Chen C. Identification of the relationship between the gut microbiome and feed efficiency in a commercial pig cohort. J Anim Sci 2021; 99:6133345. [PMID: 33570553 DOI: 10.1093/jas/skab045] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
Feed efficiency (FE) is an economically important trait in pig production. Gut microbiota plays an important role in energy harvest, nutrient metabolism, and fermentation of dietary indigestible components. Whether and which gut microbes affect FE in pigs are largely unknown. Here, a total of 208 healthy Duroc pigs were used as experimental materials. Feces and serum samples were collected at the age of 140 d. We first performed 16S rRNA gene and metagenomic sequencing analysis to investigate the relationship between the gut microbiome and porcine residual feed intake (RFI). 16S rRNA gene sequencing analysis detected 21 operational taxonomic units showing the tendency to correlation with the RFI (P < 0.01). Metagenomic sequencing further identified that the members of Clostridiales, e.g., Ruminococcus flavefaoiens, Lachnospiraceae bacterium 28-4, and Lachnospiraceae phytofermentans, were enriched in pigs with low RFI (high-FE), while 11 bacterial species including 5 Prevotella spp., especially, the Prevotella copri, had higher abundance in pigs with high RFI. Functional capacity analysis suggested that the gut microbiome of low RFI pigs had a high abundance of the pathways related to amino acid metabolism and biosynthesis, but a low abundance of the pathways associated with monosaccharide metabolism and lipopolysaccharide biosynthesis. Serum metabolome and fecal short-chain fatty acids were determined by UPLC-QTOF/MS and gas chromatography, respectively. Propionic acid in feces and the serum metabolites related to amino acid metabolism were negatively correlated with the RFI. The results from this study may provide potential gut microbial biomarkers that could be used for improving FE in pig production industry.
Collapse
Affiliation(s)
- Hui Jiang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, PR China
| | - Shaoming Fang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, PR China
| | - Hui Yang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, PR China.,College of Bioscience and Engineering, Jiangxi Agricultural University, Nanchang, PR China
| | - Congying Chen
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, PR China
| |
Collapse
|
30
|
Abstract
Resistant starch, microbiome, and precision modulation. Mounting evidence has positioned the gut microbiome as a nexus of health. Modulating its phylogenetic composition and function has become an attractive therapeutic prospect. Resistant starches (granular amylase-resistant α-glycans) are available as physicochemically and morphologically distinguishable products. Attempts to leverage resistant starch as microbiome-modifying interventions in clinical studies have yielded remarkable inter-individual variation. Consequently, their utility as a potential therapy likely depends predominantly on the selected resistant starch and the subject's baseline microbiome. The purpose of this review is to detail i) the heterogeneity of resistant starches, ii) how resistant starch is sequentially degraded and fermented by specialized gut microbes, and iii) how resistant starch interventions yield variable effects on the gut microbiome.
Collapse
Affiliation(s)
- Peter A. Dobranowski
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Alain Stintzi
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
31
|
Fagundes RR, Bourgonje AR, Saeed A, Vich Vila A, Plomp N, Blokzijl T, Sadaghian Sadabad M, von Martels JZH, van Leeuwen SS, Weersma RK, Dijkstra G, Harmsen HJM, Faber KN. Inulin-grown Faecalibacterium prausnitzii cross-feeds fructose to the human intestinal epithelium. Gut Microbes 2021; 13:1993582. [PMID: 34793284 PMCID: PMC8604389 DOI: 10.1080/19490976.2021.1993582] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/08/2021] [Accepted: 10/06/2021] [Indexed: 02/04/2023] Open
Abstract
Many chronic diseases are associated with decreased abundance of the gut commensal Faecalibacterium prausnitzii. This strict anaerobe can grow on dietary fibers, e.g., prebiotics, and produce high levels of butyrate, often associated to epithelial metabolism and health. However, little is known about other F. prausnitzii metabolites that may affect the colonic epithelium. Here, we analyzed prebiotic cross-feeding between F. prausnitzii and intestinal epithelial (Caco-2) cells in a "Human-oxygen Bacteria-anaerobic" coculture system. Inulin-grown F. prausnitzii enhanced Caco-2 viability and suppressed inflammation- and oxidative stress-marker expression. Inulin-grown F. prausnitzii produced excess butyrate and fructose, but only fructose efficiently promoted Caco-2 growth. Finally, fecal microbial taxonomy analysis (16S sequencing) from healthy volunteers (n = 255) showed the strongest positive correlation for F. prausnitzii abundance and stool fructose levels. We show that fructose, produced and accumulated in a fiber-rich colonic environment, supports colonic epithelium growth, while butyrate does not.
Collapse
Affiliation(s)
- Raphael R. Fagundes
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Arno R. Bourgonje
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ali Saeed
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Institute of Molecular Biology & Biotechnology, Bahauddin Zakariya University, Multan, Pakistan
| | - Arnau Vich Vila
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Niels Plomp
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tjasso Blokzijl
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mehdi Sadaghian Sadabad
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Julius Z. H. von Martels
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sander S. van Leeuwen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rinse K. Weersma
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hermie J. M. Harmsen
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
32
|
Briggs JA, Grondin JM, Brumer H. Communal living: glycan utilization by the human gut microbiota. Environ Microbiol 2020; 23:15-35. [PMID: 33185970 DOI: 10.1111/1462-2920.15317] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/15/2022]
Abstract
Our lower gastrointestinal tract plays host to a vast consortium of microbes, known as the human gut microbiota (HGM). The HGM thrives on a complex and diverse range of glycan structures from both dietary and host sources, the breakdown of which requires the concerted action of cohorts of carbohydrate-active enzymes (CAZymes), carbohydrate-binding proteins, and transporters. The glycan utilization profile of individual taxa, whether 'specialist' or 'generalist', is dictated by the number and functional diversity of these glycan utilization systems. Furthermore, taxa in the HGM may either compete or cooperate in glycan deconstruction, thereby creating a complex ecological web spanning diverse nutrient niches. As a result, our diet plays a central role in shaping the composition of the HGM. This review presents an overview of our current understanding of glycan utilization by the HGM on three levels: (i) molecular mechanisms of individual glycan deconstruction and uptake by key bacteria, (ii) glycan-mediated microbial interactions, and (iii) community-scale effects of dietary changes. Despite significant recent advancements, there remains much to be discovered regarding complex glycan metabolism in the HGM and its potential to affect positive health outcomes.
Collapse
Affiliation(s)
- Jonathon A Briggs
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Julie M Grondin
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Harry Brumer
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada.,Department of Chemistry, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada.,Department of Botany, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| |
Collapse
|
33
|
Abstract
Nutritional content and timing are increasingly appreciated to constitute important human variables collectively impacting all aspects of human physiology and disease. However, person-specific mechanisms driving nutritional impacts on the human host remain incompletely understood, while current dietary recommendations remain empirical and nonpersonalized. Precision nutrition aims to harness individualized bodies of data, including the human gut microbiome, in predicting person-specific physiological responses (such as glycemic responses) to food. With these advances notwithstanding, many unknowns remain, including the long-term efficacy of such interventions in delaying or reversing human metabolic disease, mechanisms driving these dietary effects, and the extent of the contribution of the gut microbiome to these processes. We summarize these conceptual advances, while highlighting challenges and means of addressing them in the next decade of study of precision medicine, toward generation of insights that may help to evolve precision nutrition as an effective future tool in a variety of "multifactorial" human disorders.
Collapse
|
34
|
Liu S, Zhao W, Liu X, Cheng L. Metagenomic analysis of the gut microbiome in atherosclerosis patients identify cross-cohort microbial signatures and potential therapeutic target. FASEB J 2020; 34:14166-14181. [PMID: 32939880 DOI: 10.1096/fj.202000622r] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/10/2020] [Accepted: 06/19/2020] [Indexed: 12/21/2022]
Abstract
The gut microbiota is associated with cardiovascular diseases, including atherosclerosis. However, the composition, functional capacity, and metabolites of the gut microbiome about atherosclerosis have not been comprehensively studied. Here, we reanalyzed 25 metagenomic stool samples from Sweden and 385 metagenomic stool samples from China using HUMAnN2, PanPhlAn, and MelonnPan to obtain more sufficient information. We found that the samples from atherosclerotic patients in both cohorts were depleted in Bacteroides xylanisolvens, Odoribacter splanchnicus, Eubacterium eligens, Roseburia inulinivorans, and Roseburia intestinalis. At the functional level, healthy metagenomes were both enriched in pathways of starch degradation V, glycolysis III (from glucose), CDP-diacylglycerol biosynthesis, and folate transformations. R inulinivorans and R intestinalis are major contributors to starch degradation V, while E eligens greatly contribute to the pathway CDP-diacylglycerol biosynthesis, and B xylanisolvens and B uniformis contribute to folate transformations II. The 11 marker species selected from the Chinese cohort distinguish patients from controls with an area under the receiver operating characteristics curve (AUC) of 0.86. Strain-level microbial analysis revealed a geographically associated adaptation of the strains from E eligens, B uniformis, and E coli. Two gut microbial metabolites, nicotinic acid and hydrocinnamic acid, had significantly higher predicted abundance in the control samples compared to the patients in the Chinese cohort, and interestinglynicotinic acid is already an effective lipid-lowering drug to reducing cardiovascular risk. Our results indicate intestinal bacteria such as B xylanisolvens, E eligens, and R inulinivorans could be promising probiotics and potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Sheng Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Department of Critical Care Medicine, Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Wenjing Zhao
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Xueyan Liu
- Department of Critical Care Medicine, Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, China
| | - Lixin Cheng
- Department of Critical Care Medicine, Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, China
| |
Collapse
|
35
|
Tarrah A, Pakroo S, Lemos Junior WJF, Guerra AF, Corich V, Giacomini A. Complete Genome Sequence and Carbohydrates-Active EnZymes (CAZymes) Analysis of Lactobacillus paracasei DTA72, a Potential Probiotic Strain with Strong Capability to Use Inulin. Curr Microbiol 2020; 77:2867-2875. [PMID: 32623485 DOI: 10.1007/s00284-020-02089-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023]
Abstract
The whole genome sequence of Lactobacillus paracasei DTA72, isolated from healthy infant feces, is reported, along with the Carbohydrates-Active enZymes (CAZymes) analysis and an in silico safety assessment. Strain DTA72 had previously demonstrated some interesting potential probiotic features, such as a good resistance to gastrointestinal conditions and an anti-Listeria activity. The 3.1 Mb sequenced genome consists of 3116 protein-coding sequences distributed on 340 SEED subsystems. In the present study, we analyzed the fermentation capability of strain DTA72 on six different carbohydrate sources, namely, glucose, fructose, lactose, galactose, xylose, and inulin by using phenotypical and genomic approaches. Interestingly, L. paracasei DTA72 evidenced the best growth performances on inulin with a much shorter lag phase and higher number of cells at the stationary phase in comparison with all the sugars tested. The CAZyme analysis using the predicted amino acid sequences detected 80 enzymes, distributed into the five CAZymes classes. Moreover, the in silico analysis revealed the absence of blood hemolytic genes, transmissible antibiotic resistances, and plasmids in DTA72. The results described in this study, together with those previously reported and particularly the strong capability to utilize inulin as energy source, make DTA72 a very interesting potential probiotic strain to be considered for the production of synbiotic foods. The complete genome data have been deposited in GenBank under the accession number WUJH00000000.
Collapse
Affiliation(s)
- Armin Tarrah
- Department of Agronomy Food Natural Resources Animal and Environment (DAFNAE), University of Padova, viale dell'Università 16, 35020, Legnaro, PD, Italy
| | - Shadi Pakroo
- Department of Agronomy Food Natural Resources Animal and Environment (DAFNAE), University of Padova, viale dell'Università 16, 35020, Legnaro, PD, Italy
| | | | - Andre Fioravante Guerra
- Department of Food Engineering, Federal Center of Technological Education Celso Suckow da Fonseca, Valença, RJ, 27.600-000, Brazil
| | - Viviana Corich
- Department of Agronomy Food Natural Resources Animal and Environment (DAFNAE), University of Padova, viale dell'Università 16, 35020, Legnaro, PD, Italy.
| | - Alessio Giacomini
- Department of Agronomy Food Natural Resources Animal and Environment (DAFNAE), University of Padova, viale dell'Università 16, 35020, Legnaro, PD, Italy
| |
Collapse
|
36
|
Logtenberg MJ, Akkerman R, An R, Hermes GDA, de Haan BJ, Faas MM, Zoetendal EG, Schols HA, de Vos P. Fermentation of Chicory Fructo-Oligosaccharides and Native Inulin by Infant Fecal Microbiota Attenuates Pro-Inflammatory Responses in Immature Dendritic Cells in an Infant-Age-Dependent and Fructan-Specific Way. Mol Nutr Food Res 2020; 64:e2000068. [PMID: 32420676 PMCID: PMC7378940 DOI: 10.1002/mnfr.202000068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/09/2020] [Indexed: 12/19/2022]
Abstract
SCOPE Inulin-type fructans are commonly applied in infant formula to support development of gut microbiota and immunity. These inulin-type fructans are considered to be fermented by gut microbiota, but it is unknown how fermentation impacts immune modulating capacity and whether the process of fermentation is dependent on the infant's age. METHODS AND RESULTS The in vitro fermentation of chicory fructo-oligosaccharides (FOS) and native inulin are investigated using pooled fecal inocula of two- and eight-week-old infants. Both inocula primarily utilize the trisaccharides in FOS, while they almost completely utilize native inulin with degree of polymerization (DP) 3-8. Fecal microbiota of eight-week-old infants degrades longer chains of native inulin up to DP 16. This correlates with a higher abundance of Bifidobacterium and higher production of acetate and lactate after 26 h of fermentation. Fermented FOS and native inulin attenuate pro-inflammatory cytokines produced by immature dendritic cells (DCs), but profiles and magnitude of attenuation are stronger with native inulin than with FOS. CONCLUSION The findings demonstrate that fermentation of FOS and native inulin is dependent on the infant's age and fructan structure. Fermentation enhances attenuating effects of pro-inflammatory responses in DCs, which depend mainly on microbial metabolites formed during fermentation.
Collapse
Affiliation(s)
- Madelon J. Logtenberg
- Laboratory of Food ChemistryWageningen University and ResearchBornse Weilanden 9, 6708 WGWageningenThe Netherlands
| | - Renate Akkerman
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical BiologyUniversity of Groningen and University Medical Centre GroningenHanzeplein 1, 9700 RBGroningenThe Netherlands
| | - Ran An
- Laboratory of MicrobiologyWageningen University and ResearchStippeneng 4, 6708 WEWageningenThe Netherlands
| | - Gerben D. A. Hermes
- Laboratory of MicrobiologyWageningen University and ResearchStippeneng 4, 6708 WEWageningenThe Netherlands
| | - Bart J. de Haan
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical BiologyUniversity of Groningen and University Medical Centre GroningenHanzeplein 1, 9700 RBGroningenThe Netherlands
| | - Marijke M. Faas
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical BiologyUniversity of Groningen and University Medical Centre GroningenHanzeplein 1, 9700 RBGroningenThe Netherlands
| | - Erwin G. Zoetendal
- Laboratory of MicrobiologyWageningen University and ResearchStippeneng 4, 6708 WEWageningenThe Netherlands
| | - Henk A. Schols
- Laboratory of Food ChemistryWageningen University and ResearchBornse Weilanden 9, 6708 WGWageningenThe Netherlands
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical BiologyUniversity of Groningen and University Medical Centre GroningenHanzeplein 1, 9700 RBGroningenThe Netherlands
| |
Collapse
|
37
|
Biasato I, Ferrocino I, Colombino E, Gai F, Schiavone A, Cocolin L, Vincenti V, Capucchio MT, Gasco L. Effects of dietary Hermetia illucens meal inclusion on cecal microbiota and small intestinal mucin dynamics and infiltration with immune cells of weaned piglets. J Anim Sci Biotechnol 2020; 11:64. [PMID: 32587688 PMCID: PMC7313100 DOI: 10.1186/s40104-020-00466-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023] Open
Abstract
Background The constant interaction between diet and intestinal barrier has a crucial role in determining gut health in pigs. Hermetia illucens (HI) meal (that represents a promising, alternative feed ingredient for production animals) has recently been demonstrated to influence colonic microbiota, bacterial metabolite profile and mucosal immune status of pigs, but no data about modulation of gut mucin dynamics are currently available. The present study evaluated the effects of dietary HI meal inclusion on the small intestinal mucin composition of piglets, as well as providing insights into the cecal microbiota and the mucosal infiltration with immune cells. Results A total of 48 weaned piglets were randomly allotted to 3 dietary treatments (control diet [C] and 5% or 10% HI meal inclusion [HI5 and HI10], with 4 replicate boxes/treatment and 4 animals/box) and slaughtered after 61 days of trial (3 animals/box, 12 piglets/diet). The cecal microbiota assessment by 16S rRNA amplicon based sequencing showed higher beta diversity in the piglets fed the HI-based diets than the C (P < 0.001). Furthermore, the HI-fed animals showed increased abundance of Blautia, Chlamydia, Coprococcus, Eubacterium, Prevotella, Roseburia, unclassified members of Ruminococcaceae, Ruminococcus and Staphylococcus when compared to the C group (FDR < 0.05). The gut of the piglets fed the HI-based diets showed greater neutral mucin percentage than the C (P < 0.05), with the intestinal neutral mucins of the HI-fed animals being also higher than the sialomucins and the sulfomucins found in the gut of the C group (P < 0.05). Furthermore, the piglets fed the HI-based diets displayed lower histological scores in the jejunum than the other gut segments (ileum [HI5] or ileum and duodenum [HI10], P < 0.05). Conclusions Dietary HI meal utilization positively influenced the cecal microbiota and the small intestinal mucin dynamics of the piglets in terms of selection of potentially beneficial bacteria and preservation of mature mucin secretory architecture, without determining the development of gut inflammation. These findings further confirm the suitability of including insect meal in swine diets.
Collapse
Affiliation(s)
- Ilaria Biasato
- Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Ilario Ferrocino
- Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Elena Colombino
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Francesco Gai
- Institute of Science of Food Production, National Research Council, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Achille Schiavone
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy.,Institute of Science of Food Production, National Research Council, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Luca Cocolin
- Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Valeria Vincenti
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Maria Teresa Capucchio
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy.,Institute of Science of Food Production, National Research Council, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Laura Gasco
- Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| |
Collapse
|
38
|
Vacca M, Celano G, Calabrese FM, Portincasa P, Gobbetti M, De Angelis M. The Controversial Role of Human Gut Lachnospiraceae. Microorganisms 2020; 8:E573. [PMID: 32326636 PMCID: PMC7232163 DOI: 10.3390/microorganisms8040573] [Citation(s) in RCA: 889] [Impact Index Per Article: 177.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/05/2020] [Accepted: 04/13/2020] [Indexed: 02/06/2023] Open
Abstract
The complex polymicrobial composition of human gut microbiota plays a key role in health and disease. Lachnospiraceae belong to the core of gut microbiota, colonizing the intestinal lumen from birth and increasing, in terms of species richness and their relative abundances during the host's life. Although, members of Lachnospiraceae are among the main producers of short-chain fatty acids, different taxa of Lachnospiraceae are also associated with different intra- and extraintestinal diseases. Their impact on the host physiology is often inconsistent across different studies. Here, we discuss changes in Lachnospiraceae abundances according to health and disease. With the aim of harnessing Lachnospiraceae to promote human health, we also analyze how nutrients from the host diet can influence their growth and how their metabolites can, in turn, influence host physiology.
Collapse
Affiliation(s)
- Mirco Vacca
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| | - Giuseppe Celano
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, 70121 Bari, Italy
| | - Marco Gobbetti
- Faculty of Science and Technology, Free University of Bozen, 39100 Bolzano, Italy;
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.V.); (F.M.C.); (M.D.A.)
| |
Collapse
|
39
|
Sheridan PO, Martin JC, Scott KP. Conjugation Protocol Optimised for Roseburia inulinivorans and Eubacterium rectale. Bio Protoc 2020; 10:e3575. [PMID: 33659545 DOI: 10.21769/bioprotoc.3575] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/17/2020] [Accepted: 03/11/2020] [Indexed: 11/02/2022] Open
Abstract
Roseburia and Eubacterium species of the human gut microbiota play an important role in the maintaince of human health, partly by producing butyrate, the main energy source of our colonic epithelial cells. However, our knowledge of the biochemistry and physiology of these bacteria has been limited by a lack of genetic manipulation techniques. Conjugative transposons previously introduced into Roseburia species could not be easily modified, greatly limiting their applicability as genetic modification platforms. Modular plasmid shuttle vectors have previously been developed for Clostridium species, which share a taxonomic order with Roseburia and Eubacterium, raising the possibility that these vectors could be used in these organisms. Here, we describe an optimized conjugation protocol enabling the transfer of autonomously replicating plasmids from an E. coli donor strain into Roseburia inulinivorans and Eubacterium rectale. The modular nature of the plasmids and their ability to be maintained in the recipient bacterium by autonomous replication makes them ideal for investigating heterologous gene expression, and as a platform for other genetic tools including antisense RNA silencing or mobile group II interon gene disruption strategies.
Collapse
Affiliation(s)
- Paul O Sheridan
- Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Jennifer C Martin
- Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Karen P Scott
- Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK
| |
Collapse
|
40
|
Abstract
Dietary fibers can be utilized to shape the human gut microbiota. However, the outcomes from most dietary fibers currently used as prebiotics are a result of competition between microbes with overlapping abilities to utilize these fibers. Thus, divergent fiber responses are observed across individuals harboring distinct microbial communities. Here, we propose that dietary fibers can be classified hierarchically according to their specificity toward gut microbes. Highly specific fibers harbor chemical and physical characteristics that allow them to be utilized by only a narrow group of bacteria within the gut, reducing competition for that substrate. The use of such fibers as prebiotics targeted to specific microbes would result in predictable shifts independent of the background microbial composition.
Collapse
|
41
|
Biasato I, Ferrocino I, Dabbou S, Evangelista R, Gai F, Gasco L, Cocolin L, Capucchio MT, Schiavone A. Black soldier fly and gut health in broiler chickens: insights into the relationship between cecal microbiota and intestinal mucin composition. J Anim Sci Biotechnol 2020; 11:11. [PMID: 32025297 PMCID: PMC6996183 DOI: 10.1186/s40104-019-0413-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/09/2019] [Indexed: 12/17/2022] Open
Abstract
Background The relationship between diet and intestinal microbiota and mucin composition appears to be fundamental for poultry gut health. The effects of insect meal (whose role as alternative feed ingredient is now well recognized) on gut microbiota and mucin composition have recently been reported in Tenebrio molitor-fed free-range and broiler chickens, but no data are currently available for Hermetia illucens (HI)-fed broilers. The present study evaluated the effects of dietary HI meal inclusion on cecal microbiota and intestinal mucin composition of broiler chickens. Results A total of 256 male broiler chickens were allotted to 4 dietary treatments (control diet [C] and 5%, 10% and 15% HI meal inclusion, with 8 replicate pens/treatment and 8 birds/pen) and slaughtered at 35 d of age (2 animals/pen, 16 birds/diet). The cecal microbiota assessment by 16S rRNA amplicon based sequencing showed lower alpha diversity in HI15 chickens (Shannon, P < 0.05) and higher beta diversity (Adonis and ANOSIM, P < 0.001) in birds fed HI diets than C. Furthermore, HI15 birds displayed significant increase of the relative abundance of Proteobacteria phylum (False Discovery Rate [FDR] < 0.05) when compared to HI10. L-Ruminococcus (Ruminococcus from Lachnospiraceae family), Faecalibacterium, Blautia and Clostridium genera were found to be characteristic of HI5 cecal microbiota (FDR < 0.05), while broiler chickens fed HI10 and HI15 diets were characterized (FDR < 0.05) by Lactobacillus and Ruminococcus (HI10) and Bacteroides, Roseburia and Helicobacter genera (HI15). Periodic-acid Schiff, Alcian Blue pH 2.5 and high iron diamine staining on small and large intestine also demonstrated lower mucin staining intensity in the intestinal villi of HI10 and HI15 birds than C (P < 0.05). Conclusions Dietary HI meal utilization at low inclusion levels (i.e., 5%) positively influenced either the cecal microbiota or the gut mucin dynamics in terms of selection of potentially beneficial bacteria and increase in villi mucins. However, high inclusion levels (in particular the 15%) may have a negative influence in terms of partial reduction of microbial complexity, reduction of potentially beneficial bacteria, selection of bacteria with mucolytic activity and decrease in villi mucins.
Collapse
Affiliation(s)
- Ilaria Biasato
- 1Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Ilario Ferrocino
- 1Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Sihem Dabbou
- 2Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Rocchina Evangelista
- 2Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Francesco Gai
- 3Institute of Science of Food Production, National Research Council, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Laura Gasco
- 1Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Luca Cocolin
- 1Department of Agricultural, Forest and Food Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Maria Teresa Capucchio
- 2Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy.,3Institute of Science of Food Production, National Research Council, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| | - Achille Schiavone
- 2Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy.,3Institute of Science of Food Production, National Research Council, Largo Paolo Braccini 2, 10095 Grugliasco, TO Italy
| |
Collapse
|
42
|
Cerqueira FM, Photenhauer AL, Pollet RM, Brown HA, Koropatkin NM. Starch Digestion by Gut Bacteria: Crowdsourcing for Carbs. Trends Microbiol 2020; 28:95-108. [DOI: 10.1016/j.tim.2019.09.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/29/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022]
|
43
|
Alcohol or Gut Microbiota: Who Is the Guilty? Int J Mol Sci 2019; 20:ijms20184568. [PMID: 31540133 PMCID: PMC6770333 DOI: 10.3390/ijms20184568] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD), a disorder caused by excessive alcohol intake represents a global health care burden. ALD encompasses a broad spectrum of hepatic injuries including asymptomatic steatosis, alcoholic steatohepatitis (ASH), fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). The susceptibility of alcoholic patients to develop ALD is highly variable and its progression to more advanced stages is strongly influenced by several hits (i.e., amount and duration of alcohol abuse). Among them, the intestinal microbiota and its metabolites have been recently identified as paramount in ALD pathophysiology. Ethanol abuse triggers qualitative and quantitative modifications in intestinal flora taxonomic composition, mucosal inflammation, and intestinal barrier derangement. Intestinal hypermeability results in the translocation of viable pathogenic bacteria, Gram-negative microbial products, and pro-inflammatory luminal metabolites into the bloodstream, further corroborating the alcohol-induced liver damage. Thus, the premise of this review is to discuss the beneficial effect of gut microbiota modulation as a novel therapeutic approach in ALD management.
Collapse
|
44
|
Sheridan PO, Martin JC, Minton NP, Flint HJ, O'Toole PW, Scott KP. Heterologous gene expression in the human gut bacteria Eubacterium rectale and Roseburia inulinivorans by means of conjugative plasmids. Anaerobe 2019; 59:131-140. [PMID: 31228669 DOI: 10.1016/j.anaerobe.2019.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/24/2019] [Accepted: 06/18/2019] [Indexed: 01/30/2023]
Abstract
Commensal butyrate-producing bacteria in the Firmicutes phylum are abundant in the human intestine and are important for maintaining health. However, understanding of the metabolism and host interaction of these bacteria is limited by the lack of genetic modification techniques. Here we establish a protocol enabling the transfer of autonomously-replicating shuttle vectors by conjugative plasmid transfer from an Escherichia coli donor into representatives of an important sub-group of strictly anaerobic human colonic Firmicutes. Five different plasmid shuttle vectors were tested, each carrying a different origin of replication from Gram-positive bacteria. Plasmid pMTL83151 (pCB102 replicon) were successfully transferred into two strains of Eubacterium rectale, while pMTL83151 and pMTL82151 (pBP1 replicon) were transferred into Roseburia inulinivorans A2-194. Plasmids that carried a Streptococcus bovis JB1 glycoside hydrolase family 16 β-(1,3-1,4)-glucanase gene were constructed and conjugated into Roseburia inulinivorans A2-194 and Eubacterium rectale T1-815, resulting in successful heterologous expression of this introduced enzymatic activity in these two strains of butyrate-producing Firmicutes.
Collapse
Affiliation(s)
- Paul O Sheridan
- Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK; School of Microbiology & APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jennifer C Martin
- Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Nigel P Minton
- Centre for Biomolecular Sciences, University Park, Nottingham, NG7 2RD, UK
| | - Harry J Flint
- Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | - Paul W O'Toole
- School of Microbiology & APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Karen P Scott
- Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
45
|
Wang J, Lang T, Shen J, Dai J, Tian L, Wang X. Core Gut Bacteria Analysis of Healthy Mice. Front Microbiol 2019; 10:887. [PMID: 31105675 PMCID: PMC6491893 DOI: 10.3389/fmicb.2019.00887] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 04/05/2019] [Indexed: 12/16/2022] Open
Abstract
Previous studies revealed that there existed great individual variations of gut microbiota in mice, and the gut bacteria of mice were changed with the occurrence and development of diseases. To identify the core gut bacteria in healthy mice and explore their relationships with the host phenotypes would help to understand the underlying mechanisms. In this study, we identified 37 genus-level core bacteria from feces of 101 healthy mice with different ages, sexes, and mouse strains in three previous studies. They collectively represented nearly half of the total sequences, and predominantly included carbohydrate- and amino acids-metabolizing bacteria and immunomodulatory bacteria. Among them, Anaerostipes indwelt the gut of all healthy mice. Co-abundance analysis showed that these core genera were clustered into five groups (Group C1–C5), which were ecologically related. For example, the abundances of Group C2 including probiotics Bifidobacterium and Lactobacillus slightly positively correlated with those of Group C1. Principal component analysis (PCA) and multivariate analysis of variance test revealed that these core gut genera were distinguished with age and sex, and also associated with their health/disease state. Linear discriminant analysis effect size (LEfSe) method showed that bacteria in Group C1 and C2/C3 increased with the age in infancy and early adulthood, and were more abundant in female mice than in male ones. The metabolic syndrome (MS) induced by high fat diet (HFD) and accelerated postnatal growth would decrease Group C2 genera, whereas probiotics intervention would reverse HFD-induced reduction of Group C2. Spearman correlation analysis indicated that the principal components based on the abundance of the 37 core genera were significantly correlated with host characteristic parameters of MS. These results demonstrated that the 37 core genera in five co-abundance groups from healthy mice were related to host phenotypes. It was indicated that these prevalent gut bacterial genera could be representative of the healthy gut microbiome in gnotobiotic animal models, and might also be candidates of probiotics and fecal microbiota transplantation.
Collapse
Affiliation(s)
- Jingjing Wang
- Shanghai Key Laboratory for Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Lang
- Shanghai Key Laboratory for Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Shen
- Ministry of Education Key Laboratory for Systems Biomedicine, Shanghai Centre for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Juanjuan Dai
- Shanghai Key Laboratory for Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Tian
- Shanghai Key Laboratory for Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xingpeng Wang
- Shanghai Key Laboratory for Pancreatic Diseases, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
46
|
Wang W, Hu H, Zijlstra RT, Zheng J, Gänzle MG. Metagenomic reconstructions of gut microbial metabolism in weanling pigs. MICROBIOME 2019; 7:48. [PMID: 30914068 PMCID: PMC6436221 DOI: 10.1186/s40168-019-0662-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/11/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND The piglets' transition from milk to solid feed induces a succession of bacterial communities, enhancing the hosts' ability to harvest energy from dietary carbohydrates. To reconstruct microbial carbohydrate metabolism in weanling pigs, this study combined 16S rRNA gene sequencing (n = 191) and shotgun metagenomics (n = 72). RESULTS Time and wheat content in feed explained most of the variation of the microbiota as assessed by 16S rRNA gene sequencing in weanling pigs. De novo metagenomic binning reconstructed 360 high-quality genomes that represented 11 prokaryotic and 1 archaeal phylum. Analysis of carbohydrate metabolism in these genomes revealed that starch fermentation is carried out by a consortium of Firmicutes expressing extracellular α-(1 → 4)-glucan branching enzyme (GH13) and Bacteroidetes expressing periplasmic neopullulanase (GH13) and α-glucosidase (GH97). Fructans were degraded by extracellular GH32 enzymes from Bacteriodetes and Lactobacillus. Lactose fermentation by β-galactosidases (GH2 and GH42) was identified in Firmicutes. In conclusion, the assembly of 360 high-quality genomes as the first metagenomic reference for swine intestinal microbiota allowed identification of key microbial contributors to degradation of starch, fructans, and lactose. CONCLUSIONS Microbial consortia that are responsible for degradation of these glycans differ substantially from the microbial consortia that degrade the same glycans in humans. Our study thus enables improvement of feeding models with higher feed efficiency and better pathogen control for weanling pigs.
Collapse
Affiliation(s)
- Weilan Wang
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 4-10 Ag/For Centre, Edmonton, Alberta, T6G 2P5, Canada
- State Key Lab of Agricultural Microbiology, Hubei Key Laboratory of Agricultural Bioinformatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Huifeng Hu
- State Key Lab of Agricultural Microbiology, Hubei Key Laboratory of Agricultural Bioinformatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Ruurd T Zijlstra
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 4-10 Ag/For Centre, Edmonton, Alberta, T6G 2P5, Canada
| | - Jinshui Zheng
- State Key Lab of Agricultural Microbiology, Hubei Key Laboratory of Agricultural Bioinformatics, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| | - Michael G Gänzle
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 4-10 Ag/For Centre, Edmonton, Alberta, T6G 2P5, Canada.
- Hubei University of Technology, College of Bioengineering and Food Science, Wuhan, People's Republic of China.
| |
Collapse
|
47
|
Kovatcheva-Datchary P, Shoaie S, Lee S, Wahlström A, Nookaew I, Hallen A, Perkins R, Nielsen J, Bäckhed F. Simplified Intestinal Microbiota to Study Microbe-Diet-Host Interactions in a Mouse Model. Cell Rep 2019; 26:3772-3783.e6. [PMID: 30917328 PMCID: PMC6444000 DOI: 10.1016/j.celrep.2019.02.090] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/22/2019] [Accepted: 02/21/2019] [Indexed: 12/16/2022] Open
Abstract
The gut microbiota can modulate human metabolism through interactions with macronutrients. However, microbiota-diet-host interactions are difficult to study because bacteria interact in complex food webs in concert with the host, and many of the bacteria are not yet characterized. To reduce the complexity, we colonize mice with a simplified intestinal microbiota (SIM) composed of ten sequenced strains isolated from the human gut with complementing pathways to metabolize dietary fibers. We feed the SIM mice one of three diets (chow [fiber rich], high-fat/high-sucrose, or zero-fat/high-sucrose diets [both low in fiber]) and investigate (1) how dietary fiber, saturated fat, and sucrose affect the abundance and transcriptome of the SIM community, (2) the effect of microbe-diet interactions on circulating metabolites, and (3) how microbiota-diet interactions affect host metabolism. Our SIM model can be used in future studies to help clarify how microbiota-diet interactions contribute to metabolic diseases.
Collapse
Affiliation(s)
- Petia Kovatcheva-Datchary
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, 41345, Sweden
| | - Saeed Shoaie
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, SE1 9RT, UK
| | - Sunjae Lee
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, SE1 9RT, UK
| | - Annika Wahlström
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, 41345, Sweden
| | - Intawat Nookaew
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, 41345, Sweden
| | - Anna Hallen
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, 41345, Sweden
| | - Rosie Perkins
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, 41345, Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, 41345, Sweden; Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, 2800, Denmark
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, 41345, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, 2200, Denmark.
| |
Collapse
|
48
|
Abstract
Since the renaissance of microbiome research in the past decade, much insight has accumulated in comprehending forces shaping the architecture and functionality of resident microorganisms in the human gut. Of the multiple host-endogenous and host-exogenous factors involved, diet emerges as a pivotal determinant of gut microbiota community structure and function. By introducing dietary signals into the nexus between the host and its microbiota, nutrition sustains homeostasis or contributes to disease susceptibility. Herein, we summarize major concepts related to the effect of dietary constituents on the gut microbiota, highlighting chief principles in the diet-microbiota crosstalk. We then discuss the health benefits and detrimental consequences that the interactions between dietary and microbial factors elicit in the host. Finally, we present the promises and challenges that arise when seeking to incorporate microbiome data in dietary planning and portray the anticipated revolution that the field of nutrition is facing upon adopting these novel concepts.
Collapse
Affiliation(s)
- Niv Zmora
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel.,Gastroenterology Unit, Tel Aviv Sourasky Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jotham Suez
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
49
|
Zhu J, Liao M, Yao Z, Liang W, Li Q, Liu J, Yang H, Ji Y, Wei W, Tan A, Liang S, Chen Y, Lin H, Zhu X, Huang S, Tian J, Tang R, Wang Q, Mo Z. Breast cancer in postmenopausal women is associated with an altered gut metagenome. MICROBIOME 2018; 6:136. [PMID: 30081953 PMCID: PMC6080540 DOI: 10.1186/s40168-018-0515-3] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 07/10/2018] [Indexed: 05/12/2023]
Abstract
BACKGROUND Increasing evidence suggests that gut microbiota play a role in the pathogenesis of breast cancer. The composition and functional capacity of gut microbiota associated with breast cancer have not been studied systematically. METHODS We performed a comprehensive shotgun metagenomic analysis of 18 premenopausal breast cancer patients, 25 premenopausal healthy controls, 44 postmenopausal breast cancer patients, and 46 postmenopausal healthy controls. RESULTS Microbial diversity was higher in breast cancer patients than in controls. Relative species abundance in gut microbiota did not differ significantly between premenopausal breast cancer patients and premenopausal controls. In contrast, relative abundance of 45 species differed significantly between postmenopausal patients and postmenopausal controls: 38 species were enriched in postmenopausal patients, including Escherichia coli, Klebsiella sp_1_1_55, Prevotella amnii, Enterococcus gallinarum, Actinomyces sp. HPA0247, Shewanella putrefaciens, and Erwinia amylovora, and 7 species were less abundant in postmenopausal patients, including Eubacterium eligens and Lactobacillus vaginalis. Acinetobacter radioresistens and Enterococcus gallinarum were positively but weakly associated with expression of high-sensitivity C-reactive protein; Shewanella putrefaciens and Erwinia amylovora were positively but weakly associated with estradiol levels. Actinomyces sp. HPA0247 negatively but weakly correlated with CD3+CD8+ T cell numbers. Further characterization of metagenome functional capacity indicated that the gut metagenomes of postmenopausal breast cancer patients were enriched in genes encoding lipopolysaccharide biosynthesis, iron complex transport system, PTS system, secretion system, and beta-oxidation. CONCLUSION The composition and functions of the gut microbial community differ between postmenopausal breast cancer patients and healthy controls. The gut microbiota may regulate or respond to host immunity and metabolic balance. Thus, while cause and effect cannot be determined, there is a reproducible change in the microbiota of treatment-naive patients relative to matched controls.
Collapse
Affiliation(s)
- Jia Zhu
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Breast Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330000, Jiangxi, China
| | - Ming Liao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ziting Yao
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wenying Liang
- Clabee Genomics, Urban Garden Building, Bookstore Road, Luohu District, Shenzhen, 518000, Guangdong, China
| | - Qibin Li
- Clabee Genomics, Urban Garden Building, Bookstore Road, Luohu District, Shenzhen, 518000, Guangdong, China
| | - Jianlun Liu
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Huawei Yang
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yinan Ji
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wei Wei
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Aihua Tan
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Chemotherapy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Siyuan Liang
- Department of Colorectal Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yang Chen
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Haisong Lin
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiujuan Zhu
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Shengzhu Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jiarong Tian
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Ruiqiang Tang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Qiuyan Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Zengnan Mo
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
50
|
Glycan Utilization and Cross-Feeding Activities by Bifidobacteria. Trends Microbiol 2018; 26:339-350. [DOI: 10.1016/j.tim.2017.10.001] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/02/2017] [Accepted: 10/12/2017] [Indexed: 01/16/2023]
|