1
|
Hoang PH, Nguyen MT, Ngo HTT, Chu NH, Ha PT, Bui HG, To LH. Enhancement of Bioactive Compounds and Survival of Lactobacillus acidophilus Grown in the Omega-6, -7 Riched Cyanobacteria Spirulina platensis. Curr Microbiol 2024; 81:380. [PMID: 39340578 DOI: 10.1007/s00284-024-03865-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024]
Abstract
Lactobacillus acidophilus is a probiotic commonly used in aquaculture to enhance the growth and immune system of aquatic species through the synthesis of various enzymes, and antimicrobial compounds like lactic acid. Traditional method of growing L. acidophilus involes using the De Man-Rogosa-Sharpe (MRS) medium. However, L. acidophilus belongs to a non-spore forming group, which make it vulnerable to stress conditions, especially during the usage process. Therefore, the present study aimed to improve the survival rate, antibacterial activity, and enrich the polyunsaturated fatty acids (PUFAs) content of L. acidophilus LB when cultured in an algae-supplemented medium, thus increasing its benefits in aquaculture applications. Using different algae biomass species as an alternative to MRS medium for the growth of L. acidophilus LB, the results showed that Spirulina platensis promoted the highest density of L. acidophilus LB. When grown in (S. platensis + glucose) medium, L. acidophilus LB produced the highest lactic acid concentration of 18.24 ± 2.43 mg/mL and survived in extreme conditions such as 4% NaCl, pH 1.0-2.0, and 50 ºC, and inhibited 99.82 ± 0.24% of Vibrio parahaemolyticus population after 2 days of treatment. Additionally, it was observed that the PUFAs content, specifically omega-6, and -7, also increased in the fermentation mixture as compared to the control sample. These findings highlighted the potential of utilizing the cyanobacteria S. platensis as an alternative, eco-friendly growth substance for L. acidophilus LB to enhance its bioactivity and viability under extreme conditions.
Collapse
Affiliation(s)
- Phuong Ha Hoang
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, 100000, Vietnam.
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, 100000, Vietnam.
| | - Minh T Nguyen
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, 100000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, 100000, Vietnam
| | - Hoai Thu T Ngo
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, 100000, Vietnam
| | - Nhat Huy Chu
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, 100000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, 100000, Vietnam
| | - Phuong Thu Ha
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, 100000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, 100000, Vietnam
| | - Huong Giang Bui
- Institute of Biotechnology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, 100000, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, 100000, Vietnam
| | - Linh Hang To
- University of Adelaide, (Adelaide) 230 North Tce, Adelaide, SA, 5005, Australia
| |
Collapse
|
2
|
Feng G, Deng M, Li R, Hou G, Ouyang Q, Jiang X, Liu X, Tang H, Chen F, Pu S, Wan D, Yin Y. Gastrointestinal microbiota and metabolites responses to dietary cereal grains in an adult pig model. Front Microbiol 2024; 15:1442077. [PMID: 39355428 PMCID: PMC11442370 DOI: 10.3389/fmicb.2024.1442077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 08/16/2024] [Indexed: 10/03/2024] Open
Abstract
Corn (C), wheat (W), and paddy rice (PR) are important energy sources and are commonly used in feed production for swine. This study mainly focuses on the variation and regularities of microbiota and metabolites in the gastrointestinal tract (GIT) of pigs in response to C, W, and PR. A total of 18 pigs were allotted into three dietary groups with six replicated pigs and received diets containing C, W, or PR as the sole energy source, respectively. The results showed that digestive parts significantly affected the diversity of microbial communities. Cereal grain sources significantly influenced the β-diversity of microbial communities in the colon and rectum. Campylobacterota and Proteobacteria are mainly distributed in the duodenum, Lactobacillus in the jejunum, and Bacteroidota in the colon and rectum. The W diet increased the Bacteroidota, Spirochaetota, and Prevotellaceae_NK3B31_group abundances and showed the highest concentrations of all short-chain fatty acids (SCFAs) in the hindgut. Fibrobacterota, Bacteroidota, Spirochaetota, Prevotellaceae_NK3B31_group, Prevotella, and Treponema in the colon or rectum were positively correlated with acetate, propionate, butyrate, and total SCFAs. These findings suggested that aerobic bacteria and facultative anaerobes in the foregut will gradually be replaced by anaerobes in the hindgut. The W diet had the best fermentability and was beneficial to the colonization of microbial communities that mainly used carbohydrates. The hindgut flora of the PR diet group may be more balanced with fewer potential pathogenic bacteria. Many microbial communities have been identified to contribute positively to the SCFA production of the hindgut. Collectively, our study revealed the spatial variation regularities of GIT microbial communities in an adult pig model and provided new insights into GIT microbiota and responses of metabolites to cereal grain diets.
Collapse
Affiliation(s)
- Ganyi Feng
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, National Engineering Laboratory for Poultry Breeding Pollution Control and Resource Technology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Menglong Deng
- College of Animal Science and Technology, Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, China
| | - Rui Li
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, National Engineering Laboratory for Poultry Breeding Pollution Control and Resource Technology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Gaifeng Hou
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, National Engineering Laboratory for Poultry Breeding Pollution Control and Resource Technology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Qing Ouyang
- College of Animal Science and Technology, Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, China
| | - Xianji Jiang
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, National Engineering Laboratory for Poultry Breeding Pollution Control and Resource Technology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- College of Animal Science and Technology, Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, China
| | - Xiaojie Liu
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, National Engineering Laboratory for Poultry Breeding Pollution Control and Resource Technology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- College of Animal Science and Technology, Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, China
| | - Hui Tang
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, National Engineering Laboratory for Poultry Breeding Pollution Control and Resource Technology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
- College of Animal Science and Technology, Hunan Co-Innovation Center of Animal Production Safety, Hunan Agricultural University, Changsha, China
| | - Fengming Chen
- Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Shihua Pu
- Chongqing Academy of Animal Science, Rongchang, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
| | - Dan Wan
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, National Engineering Laboratory for Poultry Breeding Pollution Control and Resource Technology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Hunan Research Center of Livestock and Poultry Sciences, South Central Experimental Station of Animal Nutrition and Feed Science in the Ministry of Agriculture, National Engineering Laboratory for Poultry Breeding Pollution Control and Resource Technology, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| |
Collapse
|
3
|
Yüksel E, Voragen AGJ, Kort R. The pectin metabolizing capacity of the human gut microbiota. Crit Rev Food Sci Nutr 2024:1-23. [PMID: 39264366 DOI: 10.1080/10408398.2024.2400235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
The human gastrointestinal microbiota, densely populated with a diverse array of microorganisms primarily from the bacterial phyla Bacteroidota, Bacillota, and Actinomycetota, is crucial for maintaining health and physiological functions. Dietary fibers, particularly pectin, significantly influence the composition and metabolic activity of the gut microbiome. Pectin is fermented by gut bacteria using carbohydrate-active enzymes (CAZymes), resulting in the production of short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate, which provide various health benefits. The gastrointestinal microbiota has evolved to produce CAZymes that target different pectin components, facilitating cross-feeding within the microbial community. This review explores the fermentation of pectin by various gut bacteria, focusing on the involved transport systems, CAZyme families, SCFA synthesis capacity, and effects on microbial ecology in the gut. It addresses the complexities of the gut microbiome's response to pectin and highlights the importance of microbial cross-feeding in maintaining a balanced and diverse gut ecosystem. Through a systematic analysis of pectinolytic CAZyme production, this review provides insights into the enzymatic mechanisms underlying pectin degradation and their broader implications for human health, paving the way for more targeted and personalized dietary strategies.
Collapse
Affiliation(s)
- Ecem Yüksel
- Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Alphons G J Voragen
- Keep Food Simple, Driebergen, The Netherlands
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Remco Kort
- Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- ARTIS-Micropia, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Yoon KN, Yang J, Yeom SJ, Kim SS, Park JH, Song BS, Eun JB, Park SH, Lee JH, Kim HB, Lee JH, Kim JK. Lactiplantibacillus argentoratensis AGMB00912 protects weaning mice from ETEC infection and enhances gut health. Front Microbiol 2024; 15:1440134. [PMID: 39318427 PMCID: PMC11420142 DOI: 10.3389/fmicb.2024.1440134] [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: 05/30/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Maintaining a healthy intestinal environment, optimal epithelial barrier integrity, and balanced gut microbiota composition are essential for the growth performance of weaning pigs. We identified Lactiplantibacillus argentoratensis AGMB00912 (LA) in healthy porcine feces as having antimicrobial activity against pathogens and enhanced short-chain fatty acid (SCFA) production. Herein, we assess the protective role of LA using a weaning mouse model with enterotoxigenic Escherichia coli (ETEC) infection. LA treatment improves feed intake and weight gain and alleviates colon shortening. Furthermore, LA inhibits intestinal damage, increases the small intestine villus height compared with the ETEC group, and enhances SCFA production. Using the Kyoto Encyclopedia of Genes and Genomes and other bioinformatic tools, including InterProScan and COGNIZER, we validated the presence of SCFA-producing pathways of LA and Lactiplantibacillus after whole genome sequencing. LA mitigates ETEC-induced shifts in the gut microbiota, decreasing the proportion of Escherichia and Enterococcus and increasing SCFA-producing bacteria, including Kineothrix, Lachnoclostridium, Roseuburia, Lacrimispora, Jutongia, and Blautia. Metabolic functional prediction analysis revealed enhanced functions linked to carbohydrate, amino acid, and vitamin biosynthesis, along with decreased functions associated with infectious bacterial diseases compared to the ETEC group. LA mitigates the adverse effects of ETEC infection in weaning mice, enhances growth performance and intestinal integrity, rebalances gut microbiota, and promotes beneficial metabolic functions. These findings validate the functionality of LA in a small animal model, supporting its potential application in improving the health and growth performance of weaning pigs.
Collapse
Affiliation(s)
- Ki-Nam Yoon
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, Republic of Korea
| | - Jihye Yang
- Departments of Food and Animal Biotechnology and Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Seo-Joon Yeom
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Sang-Su Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Jong-Heum Park
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Beom-Seok Song
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| | - Jong-Bang Eun
- Department of Food Science and Technology, Graduate School of Chonnam National University, Gwangju, Republic of Korea
| | - Seung-Hwan Park
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - Ju Huck Lee
- Korean Collection for Type Cultures, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Republic of Korea
| | - Hyeun Bum Kim
- Department of Animal Resources Science, Dankook University, Cheonan, Republic of Korea
| | - Ju-Hoon Lee
- Departments of Food and Animal Biotechnology and Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Jae-Kyung Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Republic of Korea
| |
Collapse
|
5
|
Jones JM, Reinke SN, Mousavi-Derazmahalleh M, Garssen J, Jenmalm MC, Srinivasjois R, Silva D, Keelan J, Prescott SL, Palmer DJ, Christophersen CT. Maternal prebiotic supplementation during pregnancy and lactation modifies the microbiome and short chain fatty acid profile of both mother and infant. Clin Nutr 2024; 43:969-980. [PMID: 38452522 DOI: 10.1016/j.clnu.2024.02.030] [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: 01/12/2024] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND & AIMS Improving maternal gut health in pregnancy and lactation is a potential strategy to improve immune and metabolic health in offspring and curtail the rising rates of inflammatory diseases linked to alterations in gut microbiota. Here, we investigate the effects of a maternal prebiotic supplement (galacto-oligosaccharides and fructo-oligosaccharides), ingested daily from <21 weeks' gestation to six months' post-partum, in a double-blinded, randomised placebo-controlled trial. METHODS Stool samples were collected at multiple timepoints from 74 mother-infant pairs as part of a larger, double-blinded, randomised controlled allergy intervention trial. The participants were randomised to one of two groups; with one group receiving 14.2 g per day of prebiotic powder (galacto-oligosaccharides GOS and fructo-oligosaccharides FOS in ratio 9:1), and the other receiving a placebo powder consisting of 8.7 g per day of maltodextrin. The faecal microbiota of both mother and infants were assessed based on the analysis of bacterial 16S rRNA gene (V4 region) sequences, and short chain fatty acid (SCFA) concentrations in stool. RESULTS Significant differences in the maternal microbiota profiles between baseline and either 28-weeks' or 36-weeks' gestation were found in the prebiotic supplemented women. Infant microbial beta-diversity also significantly differed between prebiotic and placebo groups at 12-months of age. Supplementation was associated with increased abundance of commensal Bifidobacteria in the maternal microbiota, and a reduction in the abundance of Negativicutes in both maternal and infant microbiota. There were also changes in SCFA concentrations with maternal prebiotics supplementation, including significant differences in acetic acid concentration between intervention and control groups from 20 to 28-weeks' gestation. CONCLUSION Maternal prebiotic supplementation of 14.2 g per day GOS/FOS was found to favourably modify both the maternal and the developing infant gut microbiome. These results build on our understanding of the importance of maternal diet during pregnancy, and indicate that it is possible to intervene and modify the development of the infant microbiome by dietary modulation of the maternal gut microbiome.
Collapse
Affiliation(s)
- Jacquelyn M Jones
- Trace and Environmental DNA Laboratory, School of Molecular and Life Sciences, Curtin University, Bentley, WA 6102, Australia; The Western Australian Human Microbiome Collaboration Centre, Curtin University, Bentley, WA 6027, Australia.
| | - Stacey N Reinke
- Centre for Integrative Metabolomics and Computational Biology, School of Science, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Mahsa Mousavi-Derazmahalleh
- Trace and Environmental DNA Laboratory, School of Molecular and Life Sciences, Curtin University, Bentley, WA 6102, Australia
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CS Utrecht, the Netherlands; Nutricia Research, 3584 CT Utrecht, the Netherlands
| | - Maria C Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, 581 83 Linköping, Sweden
| | - Ravisha Srinivasjois
- Joondalup Health Campus, Joondalup, WA 6027, Australia; School of Medicine, The University of Western Australia, Crawley, WA 6009, Australia
| | - Desiree Silva
- Joondalup Health Campus, Joondalup, WA 6027, Australia; School of Medicine, The University of Western Australia, Crawley, WA 6009, Australia; Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia; School of Medical & Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| | - Jeffrey Keelan
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia; School of Biomedical Sciences, The University of Western Australia, Crawley, WA 6009, Australia
| | - Susan L Prescott
- School of Medicine, The University of Western Australia, Crawley, WA 6009, Australia; Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia; Department of Immunology and Dermatology, Perth Children's Hospital, Nedlands, WA 6009, Australia; Nova Institute for Health, Baltimore, MD 21231, USA
| | - Debra J Palmer
- School of Medicine, The University of Western Australia, Crawley, WA 6009, Australia; Telethon Kids Institute, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Claus T Christophersen
- Trace and Environmental DNA Laboratory, School of Molecular and Life Sciences, Curtin University, Bentley, WA 6102, Australia; The Western Australian Human Microbiome Collaboration Centre, Curtin University, Bentley, WA 6027, Australia; Centre for Integrative Metabolomics and Computational Biology, School of Science, Edith Cowan University, Joondalup, WA 6027, Australia.
| |
Collapse
|
6
|
Toritsuka D, Aoki M, Higashida A, Fukahara K, Nishida N, Hirono K, Hane M, Sugimoto T, Asahara T, Yoshimura N. Probiotics may alleviate intestinal damage induced by cardiopulmonary bypass in children. Eur J Cardiothorac Surg 2024; 65:ezae152. [PMID: 38597892 DOI: 10.1093/ejcts/ezae152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 02/05/2024] [Accepted: 04/08/2024] [Indexed: 04/11/2024] Open
Abstract
OBJECTIVES Intestinal ischaemia-reperfusion injury induced by cardiopulmonary bypass causes intestinal epithelial barrier dysfunction, leading to dysbiosis and bacterial translocation. We conducted a randomized prospective study with 2 objectives: (i) to investigate epithelial barrier dysfunction and bacterial translocation induced by cardiopulmonary bypass and changes in the gut microbiota and (ii) to verify whether probiotics can improve these conditions. METHODS Between 2019 and 2020, patients 0-15 years old scheduled to undergo cardiac surgery using cardiopulmonary bypass were enrolled and randomly allocated to 2 groups: the intervention group received probiotics and the control group did not receive probiotics. We analysed the microbiota in faeces and blood, organic acid concentrations in faeces, plasma intestinal fatty acid-binding protein and immunological responses. RESULTS Eighty-two patients were enrolled in this study. The characteristics of the patients were similar in both groups. The total number of obligate anaerobes was higher in the intervention group than in the control group after postoperative day 7. We identified 4 clusters within the perioperative gut microbiota, and cluster changes showed a corrective effect of probiotics on dysbiosis after postoperative day 7. Organic acid concentrations in faeces, incidence of bacterial translocation, intestinal fatty acid-binding protein levels and immunological responses, except for interleukin -17A, were not markedly different between the 2 groups. CONCLUSIONS Administration of probiotics was able to correct dysbiosis but did not sufficiently alleviate the intestinal damage induced by cardiopulmonary bypass. More effective methods should be examined to prevent disturbances induced by cardiac surgery using cardiopulmonary bypass. CLINICAL TRIAL REGISTRATION NUMBER https://center6.umin.ac.jp/cgi-open-bin/ctr_e/ctr_view.cgi?recptno=R000037174 UMIN000035556.
Collapse
Affiliation(s)
- Daisuke Toritsuka
- Department of Cardiovascular Surgery, Graduate School of Medicine, University of Toyama, Toyama, Japan
| | - Masaya Aoki
- Department of Cardiovascular Surgery, Graduate School of Medicine, University of Toyama, Toyama, Japan
| | - Akihiko Higashida
- Department of Cardiovascular Surgery, Kobe Children's Hospital, Kobe, Japan
| | - Kazuaki Fukahara
- Department of Cardiovascular Surgery, Graduate School of Medicine, University of Toyama, Toyama, Japan
| | - Naonori Nishida
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, Toyama, Japan
| | - Keiichi Hirono
- Department of Pediatrics, Graduate School of Medicine, University of Toyama, Toyama, Japan
| | - Mai Hane
- Yakult Central Institute, Yakult Honsha Co., Ltd, Tokyo, Japan
| | - Takuya Sugimoto
- Yakult Central Institute, Yakult Honsha Co., Ltd, Tokyo, Japan
| | - Takashi Asahara
- Yakult Central Institute, Yakult Honsha Co., Ltd, Tokyo, Japan
| | - Naoki Yoshimura
- Department of Cardiovascular Surgery, Graduate School of Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
7
|
Muñoz-Labrador A, Kolida S, Rastall RA, Methven L, Lebrón-Aguilar R, Quintanilla-López JE, Galindo-Iranzo P, Javier Moreno F, Hernandez-Hernandez O. Prebiotic potential of new sweeteners based on the simultaneous biosynthesis of galactooligosaccharides and enzymatically modified steviol glycosides. Food Chem 2024; 436:137761. [PMID: 37862998 DOI: 10.1016/j.foodchem.2023.137761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 10/22/2023]
Abstract
Prebiotics are known for their health-promoting functions associated with the modulation of the colonic microbiota and the products of fermentation. Recently, single-pot syntheses of galactooligosaccharides in combination with steviol glycosides (mSG-GOS) have been developed. This work was conducted to evaluate their prebiotic effect by using faecal inoculum from healthy human donors during in vitro batch fermentations. Additionally, their relative sweetness was evaluated to determine their suitability as food ingredients. The results showed a significant growth (p < 0.05) of bacteria, including the genera Bifidobacterium, Bacteroides and Clostridium, and a corresponding increase in short-chain fatty acids (SCFA) in comparison to either positive and negative controls. The sweetness equivalence to 1 % w:v of SG-GOS was 0.8 % w:v when compared to sucrose. Considering the bacteria and organic acids analyses and their sweetness values of these new biosynthesized compounds, SG-GOS could act as a prebiotic sweetener with potential health benefits warranting further evaluation through human studies.
Collapse
Affiliation(s)
- Ana Muñoz-Labrador
- Institute of Food Science Research, CIAL (CSIC-UAM), C/ Nicolas Cabrera, 9, Campus Cantoblanco, 28049 Madrid, Spain.
| | - Sofia Kolida
- OptiBiotix Health plc, Innovation Centre, Innovation Way, Heslington, York YO10 5DG, UK
| | - Robert A Rastall
- Department of Food and Nutritional Sciences, The University of Reading, PO Box 226, Whiteknights, Reading RG6 6 AP, UK
| | - Lisa Methven
- Department of Food and Nutritional Sciences, The University of Reading, PO Box 226, Whiteknights, Reading RG6 6 AP, UK
| | - Rosa Lebrón-Aguilar
- Institute of Physical Chemistry "Rocasolano" (IQFR-CSIC), 28006 Madrid, Spain
| | | | | | - F Javier Moreno
- Institute of Food Science Research, CIAL (CSIC-UAM), C/ Nicolas Cabrera, 9, Campus Cantoblanco, 28049 Madrid, Spain
| | - Oswaldo Hernandez-Hernandez
- Institute of Food Science Research, CIAL (CSIC-UAM), C/ Nicolas Cabrera, 9, Campus Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
8
|
Guimarães JB, Rodrigues VF, Pereira ÍS, Manso GMDC, Elias-Oliveira J, Leite JA, Waldetario MCGM, de Oliveira S, Gomes ABDSP, Faria AMC, Ramos SG, Bonato VLD, Silva JS, Vinolo MAR, Sampaio UM, Clerici MTPS, Carlos D. Inulin prebiotic ameliorates type 1 diabetes dictating regulatory T cell homing via CCR4 to pancreatic islets and butyrogenic gut microbiota in murine model. J Leukoc Biol 2024; 115:483-496. [PMID: 37947010 DOI: 10.1093/jleuko/qiad132] [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: 11/29/2022] [Revised: 09/04/2023] [Accepted: 10/14/2023] [Indexed: 11/12/2023] Open
Abstract
Gut dysbiosis is linked to type 1 diabetes mellitus (T1D). Inulin (INU), a prebiotic, modulates the gut microbiota, promoting beneficial bacteria that produce essential short-chain fatty acids for immune regulation. However, how INU affects T1D remains uncertain. Using a streptozotocin-induced (STZ) mouse model, we studied INU's protective effects. Remarkably, STZ + INU mice resisted T1D, with none developing the disease. They had lower blood glucose, reduced pancreatic inflammation, and normalized serum insulin compared with STZ + SD mice. STZ + INU mice also had enhanced mucus production, abundant Bifidobacterium, Clostridium cluster IV, Akkermansia muciniphila, and increased fecal butyrate. In cecal lymph nodes, we observed fewer CD4+Foxp3+ regulatory T cells expressing CCR4 and more Foxp3+CCR4+ cells in pancreatic islets, with higher CCL17 expression. This phenotype was absent in CCR4-deficient mice on INU. INU supplementation effectively protects against experimental T1D by recruiting CCR4+ regulatory T cells via CCL17 into the pancreas and altering the butyrate-producing microbiota.
Collapse
Affiliation(s)
- Jhefferson Barbosa Guimarães
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Vanessa Fernandes Rodrigues
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Ítalo Sousa Pereira
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Gabriel Martins da Costa Manso
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Jefferson Elias-Oliveira
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Jefferson Antônio Leite
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | | | - Sarah de Oliveira
- Laboratory of Immunoinflammation, Department of Genetics and Evolution, Microbiology and Immunology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Arilson Bernardo Dos Santos Pereira Gomes
- Laboratory of Immunoinflammation, Department of Genetics and Evolution, Microbiology and Immunology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Ana Maria Caetano Faria
- Department of Biochemistry and Immunology, Institute of Biological Sciences, University of Minas Gerais, Belo Horizonte, Minas Gerais,31270-901, Brazil
| | - Simone Gusmão Ramos
- Laboratory of Pathology, Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Vânia L D Bonato
- Laboratory of Immunology and Pulmonary Inflammation, Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - João Santana Silva
- Fiocruz-Bi-Institutional Translational Medicine Plataform, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| | - Marco Aurélio Ramirez Vinolo
- Laboratory of Immunoinflammation, Department of Genetics and Evolution, Microbiology and Immunology, Institute of Biology, State University of Campinas, Campinas, São Paulo, Brazil
| | - Ulliana Marques Sampaio
- Department of Food Science and Nutrition, School of Food Engineering, State University of Campinas, Campinas, São Paulo, 13083-970, Brazil
| | - Maria Teresa Pedrosa Silva Clerici
- Department of Food Science and Nutrition, School of Food Engineering, State University of Campinas, Campinas, São Paulo, 13083-970, Brazil
| | - Daniela Carlos
- Laboratory of Imunorregulation of Metabolic Diseases, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ave. Bandeirantes, Ribeirão Preto, São Paulo, 14049-900, Brazil
| |
Collapse
|
9
|
Castañeda-Monsalve V, Fröhlich LF, Haange SB, Homsi MN, Rolle-Kampczyk U, Fu Q, von Bergen M, Jehmlich N. High-throughput screening of the effects of 90 xenobiotics on the simplified human gut microbiota model (SIHUMIx): a metaproteomic and metabolomic study. Front Microbiol 2024; 15:1349367. [PMID: 38444810 PMCID: PMC10912515 DOI: 10.3389/fmicb.2024.1349367] [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: 12/04/2023] [Accepted: 02/05/2024] [Indexed: 03/07/2024] Open
Abstract
The human gut microbiota is a complex microbial community with critical functions for the host, including the transformation of various chemicals. While effects on microorganisms has been evaluated using single-species models, their functional effects within more complex microbial communities remain unclear. In this study, we investigated the response of a simplified human gut microbiota model (SIHUMIx) cultivated in an in vitro bioreactor system in combination with 96 deep-well plates after exposure to 90 different xenobiotics, comprising 54 plant protection products and 36 food additives and dyes, at environmentally relevant concentrations. We employed metaproteomics and metabolomics to evaluate changes in bacterial abundances, the production of Short Chain Fatty Acids (SCFAs), and the regulation of metabolic pathways. Our findings unveiled significant changes induced by 23 out of 54 plant protection products and 28 out of 36 food additives across all three categories assessed. Notable highlights include azoxystrobin, fluroxypyr, and ethoxyquin causing a substantial reduction (log2FC < -0.5) in the concentrations of the primary SCFAs: acetate, butyrate, and propionate. Several food additives had significant effects on the relative abundances of bacterial species; for example, acid orange 7 and saccharin led to a 75% decrease in Clostridium butyricum, with saccharin causing an additional 2.5-fold increase in E. coli compared to the control. Furthermore, both groups exhibited up- and down-regulation of various pathways, including those related to the metabolism of amino acids such as histidine, valine, leucine, and isoleucine, as well as bacterial secretion systems and energy pathways like starch, sucrose, butanoate, and pyruvate metabolism. This research introduces an efficient in vitro technique that enables high-throughput screening of the structure and function of a simplified and well-defined human gut microbiota model against 90 chemicals using metaproteomics and metabolomics. We believe this approach will be instrumental in characterizing chemical-microbiota interactions especially important for regulatory chemical risk assessments.
Collapse
Affiliation(s)
- Victor Castañeda-Monsalve
- Department of Molecular Toxicology, Helmholtz Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Laura-Fabienne Fröhlich
- Department of Analytical Chemistry, Helmholtz Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Sven-Bastiaan Haange
- Department of Molecular Toxicology, Helmholtz Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Masun Nabhan Homsi
- Department of Molecular Toxicology, Helmholtz Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Toxicology, Helmholtz Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Qiuguo Fu
- Department of Analytical Chemistry, Helmholtz Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Toxicology, Helmholtz Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
- Institute of Biochemistry, Faculty of Biosciences, Pharmacy and Psychology, University of Leipzig, Leipzig, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
| | - Nico Jehmlich
- Department of Molecular Toxicology, Helmholtz Centre for Environmental Research GmbH (UFZ), Leipzig, Germany
| |
Collapse
|
10
|
Shin JH, Tillotson G, MacKenzie TN, Warren CA, Wexler HM, Goldstein EJC. Bacteroides and related species: The keystone taxa of the human gut microbiota. Anaerobe 2024; 85:102819. [PMID: 38215933 DOI: 10.1016/j.anaerobe.2024.102819] [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: 09/06/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
Microbial communities play a significant role in maintaining ecosystems in a healthy homeostasis. Presently, in the human gastrointestinal tract, there are certain taxonomic groups of importance, though there is no single species that plays a keystone role. Bacteroides spp. are known to be major players in the maintenance of eubiosis in the human gastrointestinal tract. Here we review the critical role that Bacteroides play in the human gut, their potential pathogenic role outside of the gut, and their various methods of adapting to the environment, with a focus on data for B. fragilis and B. thetaiotaomicron. Bacteroides are anaerobic non-sporing Gram negative organisms that are also resistant to bile acids, generally thriving in the gut and having a beneficial relationship with the host. While they are generally commensal organisms, some Bacteroides spp. can be opportunistic pathogens in scenarios of GI disease, trauma, cancer, or GI surgery, and cause infection, most commonly intra-abdominal infection. B. fragilis can develop antimicrobial resistance through multiple mechanisms in large part due to its plasticity and fluid genome. Bacteroidota (formerly, Bacteroidetes) have a very broad metabolic potential in the GI microbiota and can rapidly adapt their carbohydrate metabolism to the available nutrients. Gastrointestinal Bacteroidota species produce short-chain fatty acids such as succinate, acetate, butyrate, and occasionally propionate, as the major end-products, which have wide-ranging and many beneficial influences on the host. Bacteroidota, via bile acid metabolism, also play a role in in colonization-resistance of other organisms, including Clostridioides difficile, and maintenance of gut integrity.
Collapse
Affiliation(s)
- Jae Hyun Shin
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.
| | | | | | - Cirle A Warren
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.
| | - Hannah M Wexler
- GLAVAHCS, Los Angeles, CA, USA; David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | | |
Collapse
|
11
|
Gong L, Liu F, Liu J, Wang J. Dietary fiber (oligosaccharide and non-starch polysaccharide) in preventing and treating functional gastrointestinal disorders - Challenges and controversies: A review. Int J Biol Macromol 2024; 258:128835. [PMID: 38128805 DOI: 10.1016/j.ijbiomac.2023.128835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Functional gastrointestinal disorders (FGIDs) are a group of chronic or recurrent gastrointestinal functional diseases, including functional dyspepsia, irritable bowel syndrome, and functional constipation. A lack of safe and reliable treatments for abdominal pain-related FGIDs has prompted interest in new therapies. Evidence has shown that supplementation with dietary fiber may help treat FGIDs. Dietary fibers (DFs) have been demonstrated to have regulatory effects on the gut microbiota, microbiota metabolites, and gastrointestinal movement and have important implications for preventing and treating FGIDs. However, the adverse effects of some DFs, such as fermentable oligosaccharides, on FGIDs are unclear. This review provides an overview of the DFs physiological properties and functional characteristics that influence their use in management of FGIDs, with emphasis on structural modification technology to improve their therapeutic activities. The review highlights that the use of appropriate or novel fibers is a potential therapeutic approach for FGIDs.
Collapse
Affiliation(s)
- Lingxiao Gong
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Key Laboratory of Special Food Supervision Technology for State Market Regulation, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Feiyue Liu
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Key Laboratory of Special Food Supervision Technology for State Market Regulation, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Jie Liu
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Key Laboratory of Special Food Supervision Technology for State Market Regulation, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Jing Wang
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Key Laboratory of Special Food Supervision Technology for State Market Regulation, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| |
Collapse
|
12
|
Mokhtari P, Holzhausen EA, Chalifour BN, Schmidt KA, Babaei M, Machle CJ, Adise S, Alderete TL, Goran MI. Associations between Dietary Sugar and Fiber with Infant Gut Microbiome Colonization at 6 Mo of Age. J Nutr 2024; 154:152-162. [PMID: 37717629 PMCID: PMC10808822 DOI: 10.1016/j.tjnut.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/28/2023] [Accepted: 09/05/2023] [Indexed: 09/19/2023] Open
Abstract
BACKGROUND The taxonomic composition of the gut microbiome undergoes rapid development during the first 2-3 y of life. Poor diet during complementary feeding has been associated with alterations in infant growth and compromised bone, immune system, and neurodevelopment, but how it may affect gut microbial composition is unknown. OBJECTIVES This cross-sectional study aimed to examine the associations between early-life nutrition and the developing infant gut microbiota at 6 mo of age. METHODS Latino mother-infant pairs from the Mother's Milk Study (n = 105) were included. Infant gut microbiota and dietary intake were analyzed at 6 mo of age using 16S ribosomal RNA amplicon sequencing and 24-h dietary recalls, respectively. Poisson generalized linear regression analysis was performed to examine associations between dietary nutrients and microbial community abundance while adjusting for infants' mode of delivery, antibiotics, infant feeding type, time of introduction of solid foods, energy intake, and body weight. A P value of <0.05 was used to determine the statistical significance in the study. RESULTS Infants with higher consumption of total sugar exhibited a lower relative abundance of the genera Bacteroides (β = -0.01; 95% CI: -0.02, -0.00; P = 0.03) and genus Clostridium belonging to the Lachnospiraceae family (β = -0.02; 95% CI: -0.03, -0.00; P = 0.01). In addition, a higher intake of free sugar (which excludes sugar from milk, dairy, and whole fruit) was associated with several bacteria at the genus level, including Parabacteroides genus (β = 0.03; 95% CI: 0.01, 0.05; P = 0.001). Total insoluble fiber intake was associated with favorable bacteria at the genus level such as Faecalibacterium (β = 0.28; 95% CI: 0.03, 0.52; P = 0.02) and Coprococcus (β = 0.28; 95% CI: 0.02, 0.52; P = 0.03). CONCLUSION These findings demonstrate that early-life dietary intake at 6 mo impacts the developing gut microbiome associated with the presence of both unfavorable gut microbes and dietary fiber-associated commensal microbes.
Collapse
Affiliation(s)
- Pari Mokhtari
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Elizabeth A Holzhausen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Bridget N Chalifour
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Kelsey A Schmidt
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Mahsa Babaei
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Christopher J Machle
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Shana Adise
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Tanya L Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Michael I Goran
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, United States.
| |
Collapse
|
13
|
Kahleova H, Holtz DN, Strom N, La Reau A, Kolipaka S, Schmidt N, Hata E, Znayenko-Miller T, Holubkov R, Barnard ND. A dietary intervention for postmenopausal hot flashes: A potential role of gut microbiome. An exploratory analysis. Complement Ther Med 2023; 79:103002. [PMID: 37949415 DOI: 10.1016/j.ctim.2023.103002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/11/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023] Open
Abstract
OBJECTIVE This study examined the role of gut microbiome changes in mediating the effects of a dietary intervention on the frequency and severity of postmenopausal vasomotor symptoms METHODS: Postmenopausal women (n = 84) reporting ≥2 moderate-to-severe hot flashes daily were randomly assigned, in 2 successive cohorts, to an intervention including a low-fat, vegan diet and cooked soybeans (½ cup [86 g] daily) or to stay on their usual diet. Over a 12-week period, frequency and severity of hot flashes were recorded with a mobile application. In a subset of 11 women, gut microbiome was analyzed at baseline and after 12 weeks of the dietary intervention (low-fat vegan diet with soybeans), using deep shotgun metagenomic sequencing. Differences in the microbiome between baseline and 12 weeks were assessed by comparing alpha diversity with Wilcoxon signed rank tests, beta diversity with permanovaFL, and taxon abundance with Wilcoxon signed rank tests. Pearson correlations were used to assess the association between changes in hot flashes and gut bacteria. RESULTS In the subset for which microbiome testing was done, total hot flashes decreased by 95 % during the dietary intervention (p = 0.007); severe hot flashes disappeared (from 0.6 to 0.0/day; p = 0.06); and moderate-to-severe hot flashes decreased by 96 % (p = 0.01). Daytime and nighttime hot flashes were reduced by 96 % (p = 0.01) and 94 % (p = 0.004), respectively. Alpha and beta diversity did not significantly differ in the intervention group between baseline and 12 weeks. Two families (Enterobacteriaceae and Veillonellaceae), 5 genera (Erysipelatoclostridium, Fusicatenibacter, Holdemanella, Intestinimonas, and Porphyromonas), and 6 species (Clostridium asparagiforme, Clostridium innocuum, Bacteroides thetaiotaomicron, Fusicatenibacter saccharivorans, Intestinimonas butyriciproducens, Prevotella corporis, and Streptococcus sp.) were differentially abundant, but after correction for multiple comparisons, these differences were no longer significant. Changes in the relative abundance of Porphyromonas and Prevotella corporis were associated with the reduction in severe day hot flashes both unadjusted (r = 0.61; p = 0.047; and r = 0.69; p = 0.02), respectively), and after adjustment for changes in body mass index (r = 0.63; p = 0.049; and r = 0.73; p = 0.02), respectively). Changes in relative abundance of Clostridium asparagiforme were associated with the reduction in total severe hot flashes (r = 0.69; p = 0.019) and severe night hot flashes (r = 0.82; p = 0.002) and the latter association remained significant after adjustment for changes in body mass index (r = 0.75; p = 0.01). CONCLUSIONS This exploratory analysis revealed potential associations between changes in vasomotor symptoms in response to a diet change and changes in the gut microbiome. Larger randomized clinical trials are needed to investigate these findings.
Collapse
Affiliation(s)
- Hana Kahleova
- Physicians Committee for Responsible Medicine, Washington, DC, USA.
| | - Danielle N Holtz
- Physicians Committee for Responsible Medicine, Washington, DC, USA
| | | | | | - Sinjana Kolipaka
- Physicians Committee for Responsible Medicine, Washington, DC, USA; Florida Atlantic University, Charles E. Schmidt College of Medicine, FL, USA
| | - Natalie Schmidt
- Physicians Committee for Responsible Medicine, Washington, DC, USA; Florida Atlantic University, Charles E. Schmidt College of Medicine, FL, USA
| | - Ellen Hata
- Physicians Committee for Responsible Medicine, Washington, DC, USA; George Washington University, Milken Institute School of Public Health, DC, USA
| | | | | | - Neal D Barnard
- Physicians Committee for Responsible Medicine, Washington, DC, USA; Adjunct faculty, George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| |
Collapse
|
14
|
K VK, Bhat RG, Rao BK, R AP. The Gut Microbiota: a Novel Player in the Pathogenesis of Uterine Fibroids. Reprod Sci 2023; 30:3443-3455. [PMID: 37418220 PMCID: PMC10691976 DOI: 10.1007/s43032-023-01289-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/25/2023] [Indexed: 07/08/2023]
Abstract
Uterine fibroid is a common gynecological disorder that affects women of reproductive age and has emerged as a major public health concern. The symptoms have a negative influence on both their physical health and quality of life. The cost of treatment has a significant impact on the disease's burden. Even though its origin is uncertain, estrogen is thought to be a key player in fibroid pathophysiology. Many theories, including those based on genetic and environmental factors, explain what causes hyper-estrogenic condition in fibroid patients. One such possibility that is currently being explored is the hypothesis that an altered gut microbiome can contribute to the development of diseases characterized by estrogen dominance. Gut dysbiosis is often a "hot area" in the health sciences. According to a recent study, uterine fibroid patients have altered gut microbiome. A variety of risk factors influence both fibroid development and gut homeostasis. Diet, lifestyle, physical activity, and environmental contaminants have an impact on estrogen and the gut flora. A better understanding of uterine fibroids' pathophysiology is required to develop effective preventative and treatment options. A few ways by which the gut microbiota contributes to UF include estrogen, impaired immune function, inflammation, and altered gut metabolites. Therefore, in the future, while treating fibroid patients, various strategies to deal with changes in the gut flora may be advantageous. For developing suggestions for clinical diagnosis and therapy, we reviewed the literature on the relationship between uterine fibroids and the gut microbiota.
Collapse
Affiliation(s)
- Vineetha K K
- Department of Obstetrics and Gynecology, Melaka Manipal Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Rajeshwari G Bhat
- Department of Obstetrics and Gynecology, Melaka Manipal Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Bhamini Krishna Rao
- Department of Physiotherapy, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Archana P R
- Department of Basic Medical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| |
Collapse
|
15
|
Mishra BP, Mishra J, Paital B, Rath PK, Jena MK, Reddy BVV, Pati PK, Panda SK, Sahoo DK. Properties and physiological effects of dietary fiber-enriched meat products: a review. Front Nutr 2023; 10:1275341. [PMID: 38099188 PMCID: PMC10720595 DOI: 10.3389/fnut.2023.1275341] [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: 08/10/2023] [Accepted: 11/06/2023] [Indexed: 12/17/2023] Open
Abstract
Meat is a rich source of high biological proteins, vitamins, and minerals, but it is devoid of dietary fiber, an essential non-digestible carbohydrate component such as cellulose, hemicellulose, pectin, lignin, polysaccharides, and oligosaccharides. Dietary fibers are basically obtained from various cereals, legumes, fruits, vegetables, and their by-products and have numerous nutritional, functional, and health-benefiting properties. So, these fibers can be added to meat products to enhance their physicochemical properties, chemical composition, textural properties, and organoleptic qualities, as well as biological activities in controlling various lifestyle ailments such as obesity, certain cancers, type-II diabetes, cardiovascular diseases, and bowel disorders. These dietary fibers can also be used in meat products as an efficient extender/binder/filler to reduce the cost of production by increasing the cooking yield as well as by reducing the lean meat content and also as a fat replacer to minimize unhealthy fat content in the developed meat products. So, growing interest has been observed among meat processors, researchers, and scientists in exploring various new sources of dietary fibers for developing dietary fiber-enriched meat products in recent years. In the present review, various novel sources of dietary fibers, their physiological effects, their use in meat products, and their impact on various physicochemical, functional, and sensory attributes have been focused.
Collapse
Affiliation(s)
- Bidyut Prava Mishra
- Department of Livestock Products Technology, College of Veterinary Science and Animal Husbandry, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | | | - Biswaranjan Paital
- Redox Regulation Laboratory, Department of Zoology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Prasana Kumar Rath
- Department of Veterinary Pathology, College of Veterinary Science and Animal Husbandry, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | - Manoj Kumar Jena
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - B. V. Vivekananda Reddy
- Department of Livestock Products Technology, NTR College of Veterinary Science, Gannavaram, India
| | - Prasad Kumar Pati
- Department of Livestock Products Technology, College of Veterinary Science and Animal Husbandry, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | - Susen Kumar Panda
- College of Veterinary Science and Animal Husbandry, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | - Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
16
|
Albuquerque A, Garrido N, Charneca R, Egas C, Martin L, Ramos A, Costa F, Marmelo C, Martins JM. Influence of Sex and a High-Fiber Diet on the Gut Microbiome of Alentejano Pigs Raised to Heavy Weights. Vet Sci 2023; 10:641. [PMID: 37999464 PMCID: PMC10675691 DOI: 10.3390/vetsci10110641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/22/2023] [Accepted: 10/31/2023] [Indexed: 11/25/2023] Open
Abstract
This study investigates the influence of sex and a dietary transition on the gut microbiota of a local Portuguese pig breed. Three groups of male Alentejano pigs (n = 10 each) were raised between ~40 and 160 kg LW. Group C included pigs that were surgically castrated, while the I group included intact ones; both were fed with commercial diets. The third group, IExp, included intact pigs that were fed commercial diets until ~130 kg, then replaced by an experimental diet based on legumes and agro-industrial by-products between ~130 and 160 kg. Fecal samples were collected two weeks before slaughter. The total DNA was extracted and used for 16S metabarcoding on a MiSeq® System. The dietary transition from a commercial diet to the experimental diet substantially increased and shifted the diversity observed. Complex carbohydrate fermenting bacteria, such as Ruminococcus spp. and Sphaerochaeta spp., were significantly more abundant in IExp (q < 0.05). On the other hand, castrated pigs presented a significantly lower abundance of the potential probiotic, Roseburia spp. and Lachnospiraceae NK4A136 group (q < 0.01), bacteria commonly associated with better gut health and lower body fat composition. Understanding the role of gut microbiota is paramount to ensure a low skatole deposition and consumers' acceptance of pork products from non-castrated male pigs.
Collapse
Affiliation(s)
- André Albuquerque
- ECO-PIG Consortium, Z.I. Catraia, Ap. 50, 3440-131 Santa Comba Dão, Portugal; (N.G.); (R.C.); (L.M.); (A.R.); (F.C.); (C.M.)
- MED—Mediterranean Institute for Agriculture, Environment and Development & CHANGE—Global Change and Sustainability Institute, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal
| | - Nicolás Garrido
- ECO-PIG Consortium, Z.I. Catraia, Ap. 50, 3440-131 Santa Comba Dão, Portugal; (N.G.); (R.C.); (L.M.); (A.R.); (F.C.); (C.M.)
- Escola Superior Agrária de Elvas, Departamento de Ciência Agrárias e Veterinárias, Edifício Quartel do Trem, Avenida 14 de Janeiro n° 21, 7350-092 Elvas, Portugal
| | - Rui Charneca
- ECO-PIG Consortium, Z.I. Catraia, Ap. 50, 3440-131 Santa Comba Dão, Portugal; (N.G.); (R.C.); (L.M.); (A.R.); (F.C.); (C.M.)
- MED & CHANGE, Departamento de Zootecnia, ECT–Escola de Ciências e Tecnologia, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal
| | - Conceição Egas
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- Next Generation Sequencing Unit, Biocant, 3060-197 Cantanhede, Portugal
| | - Luísa Martin
- ECO-PIG Consortium, Z.I. Catraia, Ap. 50, 3440-131 Santa Comba Dão, Portugal; (N.G.); (R.C.); (L.M.); (A.R.); (F.C.); (C.M.)
- Departamento de Ciências Agrárias e Tecnologias, Escola Superior Agrária de Coimbra, Bencanta, 3045-601 Coimbra, Portugal
| | - Amélia Ramos
- ECO-PIG Consortium, Z.I. Catraia, Ap. 50, 3440-131 Santa Comba Dão, Portugal; (N.G.); (R.C.); (L.M.); (A.R.); (F.C.); (C.M.)
- Departamento de Ciências Agrárias e Tecnologias, Escola Superior Agrária de Coimbra, Bencanta, 3045-601 Coimbra, Portugal
| | - Filipa Costa
- ECO-PIG Consortium, Z.I. Catraia, Ap. 50, 3440-131 Santa Comba Dão, Portugal; (N.G.); (R.C.); (L.M.); (A.R.); (F.C.); (C.M.)
| | - Carla Marmelo
- ECO-PIG Consortium, Z.I. Catraia, Ap. 50, 3440-131 Santa Comba Dão, Portugal; (N.G.); (R.C.); (L.M.); (A.R.); (F.C.); (C.M.)
- MED—Mediterranean Institute for Agriculture, Environment and Development & CHANGE—Global Change and Sustainability Institute, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal
| | - José Manuel Martins
- ECO-PIG Consortium, Z.I. Catraia, Ap. 50, 3440-131 Santa Comba Dão, Portugal; (N.G.); (R.C.); (L.M.); (A.R.); (F.C.); (C.M.)
- MED & CHANGE, Departamento de Zootecnia, ECT–Escola de Ciências e Tecnologia, Universidade de Évora, Pólo da Mitra, Ap. 94, 7006-554 Évora, Portugal
| |
Collapse
|
17
|
Kumbhare SV, Pedroso I, Ugalde JA, Márquez-Miranda V, Sinha R, Almonacid DE. Drug and gut microbe relationships: Moving beyond antibiotics. Drug Discov Today 2023; 28:103797. [PMID: 37806386 DOI: 10.1016/j.drudis.2023.103797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/23/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
Our understanding of drug-microbe relationships has evolved from viewing microbes as mere drug producers to a dynamic, modifiable system where they can serve as drugs or targets of precision pharmacology. This review highlights recent findings on the gut microbiome, particularly focusing on four aspects of research: (i) drugs for bugs, covering recent strategies for targeting gut pathogens; (ii) bugs as drugs, including probiotics; (iii) drugs from bugs, including postbiotics; and (iv) bugs and drugs, discussing additional types of drug-microbe interactions. This review provides a perspective on future translational research, including efficient companion diagnostics in pharmaceutical interventions.
Collapse
Affiliation(s)
| | | | - Juan A Ugalde
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Valeria Márquez-Miranda
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | | | | |
Collapse
|
18
|
Guo B, Zhang J, Zhang W, Chen F, Liu B. Gut microbiota-derived short chain fatty acids act as mediators of the gut-brain axis targeting age-related neurodegenerative disorders: a narrative review. Crit Rev Food Sci Nutr 2023:1-22. [PMID: 37897083 DOI: 10.1080/10408398.2023.2272769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
Neurodegenerative diseases associated with aging are often accompanied by cognitive decline and gut microbiota disorder. But the impact of gut microbiota on these cognitive disturbances remains incompletely understood. Short chain fatty acids (SCFAs) are major metabolites produced by gut microbiota during the digestion of dietary fiber, serving as an energy source for gut epithelial cells and/or circulating to other organs, such as the liver and brain, through the bloodstream. SCFAs have been shown to cross the blood-brain barrier and played crucial roles in brain metabolism, with potential implications in mediating Alzheimer's disease (AD) and Parkinson's disease (PD). However, the underlying mechanisms that SCFAs might influence psychological functioning, including affective and cognitive processes and their neural basis, have not been fully elucidated. Furthermore, the dietary sources which determine these SCFAs production was not thoroughly evaluated yet. This comprehensive review explores the production of SCFAs by gut microbiota, their transportation through the gut-brain axis, and the potential mechanisms by which they influence age-related neurodegenerative disorders. Also, the review discusses the importance of dietary fiber sources and the challenges associated with harnessing dietary-derived SCFAs as promoters of neurological health in elderly individuals. Overall, this study suggests that gut microbiota-derived SCFAs and/or dietary fibers hold promise as potential targets and strategies for addressing age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Bingbing Guo
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Jingyi Zhang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Weihao Zhang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Feng Chen
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Innovative Development of Food Industry, Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Shenzhen University, Shenzhen, China
| | - Bin Liu
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Innovative Development of Food Industry, Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
19
|
Shin Y, Han S, Kwon J, Ju S, Choi TG, Kang I, Kim SS. Roles of Short-Chain Fatty Acids in Inflammatory Bowel Disease. Nutrients 2023; 15:4466. [PMID: 37892541 PMCID: PMC10609902 DOI: 10.3390/nu15204466] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/19/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
The gut microbiome is a diverse bacterial community in the human gastrointestinal tract that plays important roles in a variety of biological processes. Short-chain fatty acids (SCFA) are produced through fermentation of dietary fiber. Certain microbes in the gut are responsible for producing SCFAs such as acetate, propionate and butyrate. An imbalance in gut microbiome diversity can lead to metabolic disorders and inflammation-related diseases. Changes in SCFA levels and associated microbiota were observed in IBD, suggesting an association between SCFAs and disease. The gut microbiota and SCFAs affect reactive oxygen species (ROS) associated with IBD. Gut microbes and SCFAs are closely related to IBD, and it is important to study them further.
Collapse
Affiliation(s)
- Yoonhwa Shin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Juhui Kwon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Songhyun Ju
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung Soo Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (Y.S.); (S.H.); (J.K.); (S.J.)
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
20
|
Labarthe S, Plancade S, Raguideau S, Plaza Oñate F, Le Chatelier E, Leclerc M, Laroche B. Four functional profiles for fibre and mucin metabolism in the human gut microbiome. MICROBIOME 2023; 11:231. [PMID: 37858269 PMCID: PMC10588041 DOI: 10.1186/s40168-023-01667-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 09/07/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND With the emergence of metagenomic data, multiple links between the gut microbiome and the host health have been shown. Deciphering these complex interactions require evolved analysis methods focusing on the microbial ecosystem functions. Despite the fact that host or diet-derived fibres are the most abundant nutrients available in the gut, the presence of distinct functional traits regarding fibre and mucin hydrolysis, fermentation and hydrogenotrophic processes has never been investigated. RESULTS After manually selecting 91 KEGG orthologies and 33 glycoside hydrolases further aggregated in 101 functional descriptors representative of fibre and mucin degradation pathways in the gut microbiome, we used nonnegative matrix factorization to mine metagenomic datasets. Four distinct metabolic profiles were further identified on a training set of 1153 samples, thoroughly validated on a large database of 2571 unseen samples from 5 external metagenomic cohorts and confirmed with metatranscriptomic data. Profiles 1 and 2 are the main contributors to the fibre-degradation-related metagenome: they present contrasted involvement in fibre degradation and sugar metabolism and are differentially linked to dysbiosis, metabolic disease and inflammation. Profile 1 takes over Profile 2 in healthy samples, and unbalance of these profiles characterize dysbiotic samples. Furthermore, high fibre diet favours a healthy balance between profiles 1 and profile 2. Profile 3 takes over profile 2 during Crohn's disease, inducing functional reorientations towards unusual metabolism such as fucose and H2S degradation or propionate, acetone and butanediol production. Profile 4 gathers under-represented functions, like methanogenesis. Two taxonomic makes up of the profiles were investigated, using either the covariation of 203 prevalent genomes or metagenomic species, both providing consistent results in line with their functional characteristics. This taxonomic characterization showed that profiles 1 and 2 were respectively mainly composed of bacteria from the phyla Bacteroidetes and Firmicutes while profile 3 is representative of Proteobacteria and profile 4 of methanogens. CONCLUSIONS Integrating anaerobic microbiology knowledge with statistical learning can narrow down the metagenomic analysis to investigate functional profiles. Applying this approach to fibre degradation in the gut ended with 4 distinct functional profiles that can be easily monitored as markers of diet, dysbiosis, inflammation and disease. Video Abstract.
Collapse
Affiliation(s)
- Simon Labarthe
- Université Paris-Saclay, INRAE, MaIAGE, 78350, Jouy-en-Josas, France.
- Univ. Bordeaux, INRAE, BIOGECO, 33610, Cestas, France.
- Inria, INRAE, Pléiade, 33400, Talence, France.
| | - Sandra Plancade
- Université Paris-Saclay, INRAE, MaIAGE, 78350, Jouy-en-Josas, France
- UR875 MIAT, Université fédérale de Toulouse, INRAE, Castanet-Tolosan, France
| | - Sebastien Raguideau
- Université Paris-Saclay, INRAE, MaIAGE, 78350, Jouy-en-Josas, France
- Earlham Institute, Organisms and Ecosystems, NR4 7UZ, Norwich, UK
| | | | | | - Marion Leclerc
- Université Paris-Saclay, INRAE, Micalis, 78350, Jouy-en-Josas, France
- Pendulum Therapeutics, San Francisco, USA
| | - Beatrice Laroche
- Université Paris-Saclay, INRAE, MaIAGE, 78350, Jouy-en-Josas, France
- Inria, INRAE, Musca, 91120, Palaiseau, France
| |
Collapse
|
21
|
Manthei A, López-Gámez G, Martín-Belloso O, Elez-Martínez P, Soliva-Fortuny R. Relationship between Physicochemical, Techno-Functional and Health-Promoting Properties of Fiber-Rich Fruit and Vegetable By-Products and Their Enhancement by Emerging Technologies. Foods 2023; 12:3720. [PMID: 37893613 PMCID: PMC10606636 DOI: 10.3390/foods12203720] [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: 08/28/2023] [Revised: 09/20/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
The preparation and processing of fruits and vegetables produce high amounts of underutilized fractions, such as pomace and peel, which present a risk to the environment but constitute a valuable source of dietary fiber (DF) and bioactive compounds. The utilization of these fiber-rich products as functional food ingredients demands the application of treatments to improve their techno-functional properties, such as oil and water binding, and health-related properties, such as fermentability, adsorption, and retardation capacities of glucose, cholesterol, and bile acids. The enhancement of health-promoting properties is strongly connected with certain structural and techno-functional characteristics, such as the soluble DF content, presence of hydrophobic groups, and viscosity. Novel physical, environmentally friendly technologies, such as ultrasound (US), high-pressure processing (HPP), extrusion, and microwave, have been found to have higher potential than chemical and comminution techniques in causing desirable structural alterations of the DF network that lead to the improvement of techno-functionality and health promotion. The application of enzymes was related to higher soluble DF content, which might be associated with improved DF properties. Combined physical and enzymatic treatments can aid solubilization and modifications, but their benefit needs to be evaluated for each DF source and the desired outcome.
Collapse
Affiliation(s)
| | | | | | | | - Robert Soliva-Fortuny
- Department of Food Technology, Engineering and Science, University of Lleida/Agrotecnio-CeRCA Center, Av. Alcalde Rovira Roure, 191, 25198 Lleida, Spain; (A.M.)
| |
Collapse
|
22
|
Pedroza Matute S, Iyavoo S. Exploring the gut microbiota: lifestyle choices, disease associations, and personal genomics. Front Nutr 2023; 10:1225120. [PMID: 37867494 PMCID: PMC10585655 DOI: 10.3389/fnut.2023.1225120] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023] Open
Abstract
The gut microbiota is a rich and dynamic ecosystem that actively interacts with the human body, playing a significant role in the state of health and disease of the host. Diet, exercise, mental health, and other factors have exhibited the ability to influence the gut bacterial composition, leading to changes that can prevent and improve, or favor and worsen, both intestinal and extra-intestinal conditions. Altered gut microbial states, or 'dysbiosis', associated with conditions and diseases are often characterized by shifts in bacterial abundance and diversity, including an impaired Firmicutes to Bacteroidetes ratio. By understanding the effect of lifestyle on the gut microbiota, personalized advice can be generated to suit each individual profile and foster the adoption of lifestyle changes that can both prevent and ameliorate dysbiosis. The delivery of effective and reliable advice, however, depends not only on the available research and current understanding of the topic, but also on the methods used to assess individuals and to discover the associations, which can introduce bias at multiple stages. The aim of this review is to summarize how human gut microbial variability is defined and what lifestyle choices and diseases have shown association with gut bacterial composition. Furthermore, popular methods to investigate the human gut microbiota are outlined, with a focus on the possible bias caused by the lack of use of standardized methods. Finally, an overview of the current state of personalized advice based on gut microbiota testing is presented, underlining its power and limitations.
Collapse
Affiliation(s)
| | - Sasitaran Iyavoo
- Nkaarco Diagnostics Limited, Norwich, United Kingdom
- School of Chemistry, College of Health and Science, University of Lincoln, Lincoln, United Kingdom
| |
Collapse
|
23
|
Du B, Fu Y, Han Y, Sun Q, Xu J, Yang Y, Rong R. The lung-gut crosstalk in respiratory and inflammatory bowel disease. Front Cell Infect Microbiol 2023; 13:1218565. [PMID: 37680747 PMCID: PMC10482113 DOI: 10.3389/fcimb.2023.1218565] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 07/28/2023] [Indexed: 09/09/2023] Open
Abstract
Both lung and gut belong to the common mucosal immune system (CMIS), with huge surface areas exposed to the external environment. They are the main defense organs against the invasion of pathogens and play a key role in innate and adaptive immunity. Recently, more and more evidence showed that stimulation of one organ can affect the other, as exemplified by intestinal complications during respiratory disease and vice versa, which is called lung-gut crosstalk. Intestinal microbiota plays an important role in respiratory and intestinal diseases. It is known that intestinal microbial imbalance is related to inflammatory bowel disease (IBD), this imbalance could impact the integrity of the intestinal epithelial barrier and leads to the persistence of inflammation, however, gut microbial disturbances have also been observed in respiratory diseases such as asthma, allergy, chronic obstructive pulmonary disease (COPD), and respiratory infection. It is not fully clarified how these disorders happened. In this review, we summarized the latest examples and possible mechanisms of lung-gut crosstalk in respiratory disease and IBD and discussed the strategy of shaping intestinal flora to treat respiratory diseases.
Collapse
Affiliation(s)
- Baoxiang Du
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Fu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuxiu Han
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qihui Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinke Xu
- Shandong Center for Disease Control and Prevention, Jinan, China
| | - Yong Yang
- Shandong Antiviral Engineering Research Center of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Rong Rong
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
24
|
Schropp N, Stanislas V, Michels KB, Thriene K. How Do Prebiotics Affect Human Intestinal Bacteria?-Assessment of Bacterial Growth with Inulin and XOS In Vitro. Int J Mol Sci 2023; 24:12796. [PMID: 37628977 PMCID: PMC10454692 DOI: 10.3390/ijms241612796] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Prebiotics are believed to exhibit high specificity in stimulating the growth or activity of a limited number of commensal microorganisms, thereby conferring health benefits to the host. However, the mechanism of action of prebiotics depends on multiple factors, including the composition of an individual's gut microbiota, and is therefore difficult to predict. It is known that different bacteria can utilize inulin and xylooligosaccharides (XOS), but an overview of which bacteria in the human gut may be affected is lacking. Detailed knowledge of how bacterial growth is affected by prebiotics is furthermore useful for the development of new synbiotics, which combine a living microorganism with a selective substrate to confer a health benefit to the host. Hence, we developed a statistical model to compare growth in vitro among typical human gut bacteria from different phylogenetic lineages. Based on continuous observation of the optical density (OD600), we compare maximal growth rates (rmax), maximal attained OD600 (ODmax), and area under the growth curve (AUC) of bacteria grown on inulin or XOS. The consideration of these three parameters suggests strain-specific preferences for inulin or XOS and reveals previously unknown preferences such as Streptococcus salivarius growth on XOS.
Collapse
Affiliation(s)
| | | | | | - Kerstin Thriene
- Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, 79110 Freiburg, Germany; (N.S.); (V.S.); (K.B.M.)
| |
Collapse
|
25
|
Horrocks V, King OG, Yip AYG, Marques IM, McDonald JAK. Role of the gut microbiota in nutrient competition and protection against intestinal pathogen colonization. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001377. [PMID: 37540126 PMCID: PMC10482380 DOI: 10.1099/mic.0.001377] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/25/2023] [Indexed: 08/05/2023]
Abstract
The human gut microbiota can restrict the growth of pathogens to prevent them from colonizing the intestine ('colonization resistance'). However, antibiotic treatment can kill members of the gut microbiota ('gut commensals') and reduce competition for nutrients, making these nutrients available to support the growth of pathogens. This disturbance can lead to the growth and expansion of pathogens within the intestine (including antibiotic-resistant pathogens), where these pathogens can exploit the absence of competitors and the nutrient-enriched gut environment. In this review, we discuss nutrient competition between the gut microbiota and pathogens. We also provide an overview of how nutrient competition can be harnessed to support the design of next-generation microbiome therapeutics to restrict the growth of pathogens and prevent the development of invasive infections.
Collapse
Affiliation(s)
- Victoria Horrocks
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Olivia G. King
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2AZ, UK
| | - Alexander Y. G. Yip
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Inês Melo Marques
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Julie A. K. McDonald
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
26
|
Yu T, Yang W, Yao S, Yu Y, Wakamiya M, Golovko G, Cong Y. STING Promotes Intestinal IgA Production by Regulating Acetate-producing Bacteria to Maintain Host-microbiota Mutualism. Inflamm Bowel Dis 2023; 29:946-959. [PMID: 36661414 PMCID: PMC10233729 DOI: 10.1093/ibd/izac268] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Intestinal Immunoglobulin A (IgA) is crucial in maintaining host-microbiota mutualism and gut homeostasis. It has been shown that many species of gut bacteria produce cyclic dinucleotides, along with an abundance of microbiota-derived DNA present within the intestinal lumen, which triggers the tonic activation of the cytosolic cGAS-STING pathway. However, the role of STING in intestinal IgA remains poorly understood. We further investigated whether and how STING affects intestinal IgA response. METHODS Intestinal IgA was determined between wild-type (WT) mice and Sting-/- mice in steady conditions and upon enteric Citrobacter rodentium infection. STING agonists were used to stimulating B cells or dendritic cells in vitro. Gut microbiota composition was examined by 16S ribosomal RNA gene sequencing. Bacteria metabolomics functional analyses was performed by PICRUSt2. Fecal short-chain fatty acid (SCFA) was determined by Mass spectrometry and Cedex Bio Analyzer. Gut bacteria from WT mice and Sting-/- mice were transferred into germ-free mice and antibiotic-pretreated mice. RESULTS Intestinal IgA response was impaired in Sting-/- mice. However, STING agonists did not directly stimulate B cells or dendritic cells to induce IgA. Interestingly, Sting-/- mice displayed altered gut microbiota composition with decreased SCFA-producing bacteria and downregulated SCFA fermentation pathways. Transfer of fecal bacteria from Sting-/- mice induced less IgA than that from WT mice in germ-free mice and antibiotic-pretreated mice, which is mediated by GPR43. Acetate, the dominant SCFA, was decreased in Sting-/- mice, and supplementation of acetate restored intestinal IgA production in Sting-/- mice. CONCLUSIONS STING promotes intestinal IgA by regulating acetate-producing gut bacteria.
Collapse
Affiliation(s)
- Tianming Yu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Center for Microbiome Research, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Wenjing Yang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Center for Microbiome Research, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Suxia Yao
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Center for Microbiome Research, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Yanbo Yu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Maki Wakamiya
- Germ-free Mouse Facility, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - George Golovko
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Yingzi Cong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Center for Microbiome Research, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
27
|
Puljiz Z, Kumric M, Vrdoljak J, Martinovic D, Ticinovic Kurir T, Krnic MO, Urlic H, Puljiz Z, Zucko J, Dumanic P, Mikolasevic I, Bozic J. Obesity, Gut Microbiota, and Metabolome: From Pathophysiology to Nutritional Interventions. Nutrients 2023; 15:nu15102236. [PMID: 37242119 DOI: 10.3390/nu15102236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/29/2023] [Accepted: 05/07/2023] [Indexed: 05/28/2023] Open
Abstract
Obesity is a disorder identified by an inappropriate increase in weight in relation to height and is considered by many international health institutions to be a major pandemic of the 21st century. The gut microbial ecosystem impacts obesity in multiple ways that yield downstream metabolic consequences, such as affecting systemic inflammation, immune response, and energy harvest, but also the gut-host interface. Metabolomics, a systematized study of low-molecular-weight molecules that take part in metabolic pathways, represents a serviceable method for elucidation of the crosstalk between hosts' metabolism and gut microbiota. In the present review, we confer about clinical and preclinical studies exploring the association of obesity and related metabolic disorders with various gut microbiome profiles, and the effects of several dietary interventions on gut microbiome composition and the metabolome. It is well established that various nutritional interventions may serve as an efficient therapeutic approach to support weight loss in obese individuals, yet no agreement exists in regard to the most effective dietary protocol, both in the short and long term. However, metabolite profiling and the gut microbiota composition might represent an opportunity to methodically establish predictors for obesity control that are relatively simple to measure in comparison to traditional approaches, and it may also present a tool to determine the optimal nutritional intervention to ameliorate obesity in an individual. Nevertheless, a lack of adequately powered randomized trials impedes the application of observations to clinical practice.
Collapse
Affiliation(s)
- Zivana Puljiz
- Laboratory for Bioinformatics, Faculty of Food Technology and Biotechnology, University of Zagreb, 10000 Zagreb, Croatia
| | - Marko Kumric
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
| | - Josip Vrdoljak
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
| | - Dinko Martinovic
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
| | - Tina Ticinovic Kurir
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Hospital of Split, 21000 Split, Croatia
| | - Marin Ozren Krnic
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
| | - Hrvoje Urlic
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
| | - Zeljko Puljiz
- Department of Internal Medicine, University of Split School of Medicine, 21000 Split, Croatia
- Department of Gastroenterology and Hepatology, University Hospital of Split, 21000 Split, Croatia
| | - Jurica Zucko
- Laboratory for Bioinformatics, Faculty of Food Technology and Biotechnology, University of Zagreb, 10000 Zagreb, Croatia
| | - Petra Dumanic
- Medical Laboratory Diagnostic Division, University Hospital of Split, 21000 Split, Croatia
| | - Ivana Mikolasevic
- Department of Gastroenterology and Hepatology, University Hospital Centre Rijeka, School of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Josko Bozic
- Department of Pathophysiology, University of Split School of Medicine, 21000 Split, Croatia
| |
Collapse
|
28
|
Szabó C, Kachungwa Lugata J, Ortega ADSV. Gut Health and Influencing Factors in Pigs. Animals (Basel) 2023; 13:ani13081350. [PMID: 37106913 PMCID: PMC10135089 DOI: 10.3390/ani13081350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
The gastrointestinal tract (GIT) is a complex, dynamic, and critical part of the body, which plays an important role in the digestion and absorption of ingested nutrients and excreting waste products of digestion. In addition, GIT also plays a vital role in preventing the entry of harmful substances and potential pathogens into the bloodstream. The gastrointestinal tract hosts a significant number of microbes, which throughout their metabolites, directly interact with the hosts. In modern intensive animal farming, many factors can disrupt GIT functions. As dietary nutrients and biologically active substances play important roles in maintaining homeostasis and eubiosis in the GIT, this review aims to summarize the current status of our knowledge on the most important areas.
Collapse
Affiliation(s)
- Csaba Szabó
- Department of Animal Nutrition and Physiology, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
| | - James Kachungwa Lugata
- Department of Animal Nutrition and Physiology, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
- Doctoral School of Animal Science, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
| | - Arth David Sol Valmoria Ortega
- Department of Animal Nutrition and Physiology, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
- Doctoral School of Animal Science, Faculty of Agriculture and Food Sciences and Environmental Management, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary
| |
Collapse
|
29
|
Cardilli A, Hamad I, Dyczko A, Thijs S, Vangronsveld J, Müller DN, Rosshart SP, Kleinewietfeld M. Impact of High Salt-Intake on a Natural Gut Ecosystem in Wildling Mice. Nutrients 2023; 15:nu15071565. [PMID: 37049406 PMCID: PMC10096756 DOI: 10.3390/nu15071565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/28/2023] Open
Abstract
The mammalian holobiont harbors a complex and interdependent mutualistic gut bacterial community. Shifts in the composition of this bacterial consortium are known to be a key element in host health, immunity and disease. Among many others, dietary habits are impactful drivers for a potential disruption of the bacteria–host mutualistic interaction. In this context, we previously demonstrated that a high-salt diet (HSD) leads to a dysbiotic condition of murine gut microbiota, characterized by a decrease or depletion of well-known health-promoting gut bacteria. However, due to a controlled and sanitized environment, conventional laboratory mice (CLM) possess a less diverse gut microbiota compared to wild mice, leading to poor translational outcome for gut microbiome studies, since a reduced gut microbiota diversity could fail to depict the complex interdependent networks of the microbiome. Here, we evaluated the HSD effect on gut microbiota in CLM in comparison to wildling mice, which harbor a natural gut ecosystem more closely mimicking the situation in humans. Mice were treated with either control food or HSD and gut microbiota were profiled using amplicon-based methods targeting the 16S ribosomal gene. In line with previous findings, our results revealed that HSD induced significant loss of alpha diversity and extensive modulation of gut microbiota composition in CLM, characterized by the decrease in potentially beneficial bacteria from Firmicutes phylum such as the genera Lactobacillus, Roseburia, Tuzzerella, Anaerovorax and increase in Akkermansia and Parasutterella. However, HSD-treated wildling mice did not show the same changes in terms of alpha diversity and loss of Firmicutes bacteria as CLM, and more generally, wildlings exhibited only minor shifts in the gut microbiota composition upon HSD. In line with this, 16S-based functional analysis suggested only major shifts of gut microbiota ecological functions in CLM compared to wildling mice upon HSD. Our findings indicate that richer and wild-derived gut microbiota is more resistant to dietary interventions such as HSD, compared to gut microbiota of CLM, which may have important implications for future translational microbiome research.
Collapse
|
30
|
Modulation of the Tumor Microenvironment by Microbiota-Derived Short-Chain Fatty Acids: Impact in Colorectal Cancer Therapy. Int J Mol Sci 2023; 24:ijms24065069. [PMID: 36982144 PMCID: PMC10048801 DOI: 10.3390/ijms24065069] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Finding new therapeutic approaches towards colorectal cancer (CRC) is of increased relevance, as CRC is one of the most common cancers worldwide. CRC standard therapy includes surgery, chemotherapy, and radiotherapy, which may be used alone or in combination. The reported side effects and acquired resistance associated with these strategies lead to an increasing need to search for new therapies with better efficacy and less toxicity. Several studies have demonstrated the antitumorigenic properties of microbiota-derived short-chain fatty acids (SCFAs). The tumor microenvironment is composed by non-cellular components, microbiota, and a great diversity of cells, such as immune cells. The influence of SCFAs on the different constituents of the tumor microenvironment is an important issue that should be taken into consideration, and to the best of our knowledge there is a lack of reviews on this subject. The tumor microenvironment is not only closely related to the growth and development of CRC but also affects the treatment and prognosis of the patients. Immunotherapy has emerged as a new hope, but, in CRC, it was found that only a small percentage of patients benefit from this treatment being closely dependent on the genetic background of the tumors. The aim of this review was to perform an up-to-date critical literature review on current knowledge regarding the effects of microbiota-derived SCFAs in the tumor microenvironment, particularly in the context of CRC and its impact in CRC therapeutic strategies. SCFAs, namely acetate, butyrate, and propionate, have the ability to modulate the tumor microenvironment in distinct ways. SCFAs promote immune cell differentiation, downregulate the expression of pro-inflammatory mediators, and restrict the tumor-induced angiogenesis. SCFAs also sustain the integrity of basement membranes and modulate the intestinal pH. CRC patients have lower concentrations of SCFAs than healthy individuals. Increasing the production of SCFAs through the manipulation of the gut microbiota could constitute an important therapeutic strategy towards CRC due to their antitumorigenic effect and ability of modulating tumor microenvironment.
Collapse
|
31
|
Guimarães VHD, Marinho BM, Motta-Santos D, Mendes GDRL, Santos SHS. Nutritional implications in the mechanistic link between the intestinal microbiome, renin-angiotensin system, and the development of obesity and metabolic syndrome. J Nutr Biochem 2023; 113:109252. [PMID: 36509338 DOI: 10.1016/j.jnutbio.2022.109252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/12/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
Obesity and metabolic disorders represent a significant global health problem and the gut microbiota plays an important role in modulating systemic homeostasis. Recent evidence shows that microbiota and its signaling pathways may affect the whole metabolism and the Renin-Angiotensin System (RAS), which in turn seems to modify microbiota. The present review aimed to investigate nutritional implications in the mechanistic link between the intestinal microbiome, renin-angiotensin system, and the development of obesity and metabolic syndrome components. A description of metabolic changes was obtained based on relevant scientific literature. The molecular and physiological mechanisms that impact the human microbiome were addressed, including the gut microbiota associated with obesity, diabetes, and hepatic steatosis. The RAS interaction signaling and modulation were analyzed. Strategies including the use of prebiotics, symbiotics, probiotics, and biotechnology may affect the gut microbiota and its impact on human health.
Collapse
Affiliation(s)
- Victor Hugo Dantas Guimarães
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil
| | - Barbhara Mota Marinho
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil
| | - Daisy Motta-Santos
- School of Physical Education, Physiotherapy, and Occupational Therapy - EEFFTO, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Minas Gerais, Brazil
| | - Gabriela da Rocha Lemos Mendes
- Food Engineering, Institute of Agricultural Sciences (ICA), Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil
| | - Sérgio Henrique Sousa Santos
- Laboratory of Health Science, Postgraduate Program in Health Science, Universidade Estadual de Montes Claros (Unimontes), Montes Claros, Minas Gerais, Brazil; Food Engineering, Institute of Agricultural Sciences (ICA), Universidade Federal de Minas Gerais (UFMG), Montes Claros, Minas Gerais, Brazil.
| |
Collapse
|
32
|
Feng Y, Feng X, Liu S, Zhang H, Wang J. Effects of dietary fiber and ferulic acid on dough characteristics and glutenin macropolymer (GMP) aggregation behavior during dough resting. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
33
|
Muñoz-Labrador A, Lebrón-Aguilar R, Quintanilla-López JE, Galindo-Iranzo P, Azcarate SM, Kolida S, Kachrimanidou V, Garcia-Cañas V, Methven L, Rastall RA, Moreno FJ, Hernandez-Hernandez O. Prebiotic Potential of a New Sweetener Based on Galactooligosaccharides and Modified Mogrosides. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:9048-9056. [PMID: 35830712 PMCID: PMC9335866 DOI: 10.1021/acs.jafc.2c01363] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/27/2022] [Accepted: 07/02/2022] [Indexed: 06/15/2023]
Abstract
This study was conducted to investigate the sweetness intensity and the potential fecal microbiome modulation of galactooligosaccharides in combination with enzymatically modified mogrosides (mMV-GOS), both generated through a patented single-pot synthesis. Sweetness intensity was performed in vivo by trained sensory panelists. The impact on the human fecal microbiome was evaluated by in vitro pH-controlled batch fermentation, and bacterial populations and organic acid concentrations were measured by qPCR and GC-FID, respectively. Significant growth (p ≤ 0.05) during the fermentation at 10 h of bacterial populations includes Bifidobacterium (8.49 ± 0.44 CFU/mL), Bacteroides (9.73 ± 0.32 CFU/mL), Enterococcus (8.17 ± 0.42 CFU/mL), and Clostridium coccoides (6.15 ± 0.11 CFU/mL) as compared to the negative control counts for each bacterial group (7.94 ± 0.27, 7.84 ± 1.11, 7.52 ± 0.37, and 5.81 ± 0.08 CFU/mL, respectively) at the same time of fermentation. Likewise, the corresponding significant increase in production of SCFA in mMV-GOS at 10 h of fermentation, mainly seen in acetate (20.32 ± 2.56 mM) and propionate (9.49 ± 1.44 mM) production compared to a negative control at the same time (8.15 ± 1.97 and 1.86 ± 0.24 mM), is in line with a positive control (short-chain fructooligosaccharides; 46.74 ± 12.13 and 6.51 ± 1.91 mM, respectively) revealing a selective fermentation. In conclusion, these substrates could be considered as novel candidate prebiotic sweeteners, foreseeing a feasible and innovative approach targeting the sucrose content reduction in food. This new ingredient could provide health benefits when evaluated in human studies by combining sweetness and prebiotic fiber functionality.
Collapse
Affiliation(s)
- Ana Muñoz-Labrador
- Institute
of Food Science Research, CIAL (CSIC-UAM), Nicolas Cabrera, 9, 28049 Madrid, Spain
| | - Rosa Lebrón-Aguilar
- Institute
of Physical Chemistry “Rocasolano” (IQFR-CSIC), Serrano 119, 28006 Madrid, Spain
| | | | - Plácido Galindo-Iranzo
- Institute
of Physical Chemistry “Rocasolano” (IQFR-CSIC), Serrano 119, 28006 Madrid, Spain
| | - Silvana M. Azcarate
- Institute
of Earth and Environmental Sciences of La Pampa (INCITAP), Mendoza 109, L6302EPA Santa Rosa, La Pampa, Argentina
| | - Sofia Kolida
- OptiBiotix
Health Plc, Innovation Centre, Innovation Way,
Heslington, York YO10 5DG, U.K.
| | - Vasiliki Kachrimanidou
- Department
of Food and Nutritional Sciences, The University
of Reading, PO Box 226,
Whiteknights, Reading RG6 6 AP, U.K.
| | - Virginia Garcia-Cañas
- Institute
of Food Science Research, CIAL (CSIC-UAM), Nicolas Cabrera, 9, 28049 Madrid, Spain
| | - Lisa Methven
- Department
of Food and Nutritional Sciences, The University
of Reading, PO Box 226,
Whiteknights, Reading RG6 6 AP, U.K.
| | - Robert A. Rastall
- Department
of Food and Nutritional Sciences, The University
of Reading, PO Box 226,
Whiteknights, Reading RG6 6 AP, U.K.
| | - F. Javier Moreno
- Institute
of Food Science Research, CIAL (CSIC-UAM), Nicolas Cabrera, 9, 28049 Madrid, Spain
| | | |
Collapse
|
34
|
Vasquez R, Oh JK, Song JH, Kang DK. Gut microbiome-produced metabolites in pigs: a review on their biological functions and the influence of probiotics. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:671-695. [PMID: 35969697 PMCID: PMC9353353 DOI: 10.5187/jast.2022.e58] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/23/2022] [Accepted: 07/04/2022] [Indexed: 11/20/2022]
Abstract
The gastrointestinal tract is a complex ecosystem that contains a large number of microorganisms with different metabolic capacities. Modulation of the gut microbiome can improve the growth and promote health in pigs. Crosstalk between the host, diet, and the gut microbiome can influence the health of the host, potentially through the production of several metabolites with various functions. Short-chain and branched-chain fatty acids, secondary bile acids, polyamines, indoles, and phenolic compounds are metabolites produced by the gut microbiome. The gut microbiome can also produce neurotransmitters (such as γ-aminobutyric acid, catecholamines, and serotonin), their precursors, and vitamins. Several studies in pigs have demonstrated the importance of the gut microbiome and its metabolites in improving growth performance and feed efficiency, alleviating stress, and providing protection from pathogens. The use of probiotics is one of the strategies employed to target the gut microbiome of pigs. Promising results have been published on the use of probiotics in optimizing pig production. This review focuses on the role of gut microbiome-derived metabolites in the performance of pigs and the effects of probiotics on altering the levels of these metabolites.
Collapse
Affiliation(s)
- Robie Vasquez
- Department of Animal Resources Science,
Dankook University, Cheonan 31116, Korea
| | - Ju Kyoung Oh
- Department of Animal Resources Science,
Dankook University, Cheonan 31116, Korea
| | - Ji Hoon Song
- Department of Animal Resources Science,
Dankook University, Cheonan 31116, Korea
| | - Dae-Kyung Kang
- Department of Animal Resources Science,
Dankook University, Cheonan 31116, Korea
| |
Collapse
|
35
|
Lancaster SM, Lee-McMullen B, Abbott CW, Quijada JV, Hornburg D, Park H, Perelman D, Peterson DJ, Tang M, Robinson A, Ahadi S, Contrepois K, Hung CJ, Ashland M, McLaughlin T, Boonyanit A, Horning A, Sonnenburg JL, Snyder MP. Global, distinctive, and personal changes in molecular and microbial profiles by specific fibers in humans. Cell Host Microbe 2022; 30:848-862.e7. [PMID: 35483363 PMCID: PMC9187607 DOI: 10.1016/j.chom.2022.03.036] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/19/2022] [Accepted: 03/25/2022] [Indexed: 12/11/2022]
Abstract
Dietary fibers act through the microbiome to improve cardiovascular health and prevent metabolic disorders and cancer. To understand the health benefits of dietary fiber supplementation, we investigated two popular purified fibers, arabinoxylan (AX) and long-chain inulin (LCI), and a mixture of five fibers. We present multiomic signatures of metabolomics, lipidomics, proteomics, metagenomics, a cytokine panel, and clinical measurements on healthy and insulin-resistant participants. Each fiber is associated with fiber-dependent biochemical and microbial responses. AX consumption associates with a significant reduction in LDL and an increase in bile acids, contributing to its observed cholesterol reduction. LCI is associated with an increase in Bifidobacterium. However, at the highest LCI dose, there is increased inflammation and elevation in the liver enzyme alanine aminotransferase. This study yields insights into the effects of fiber supplementation and the mechanisms behind fiber-induced cholesterol reduction, and it shows effects of individual, purified fibers on the microbiome.
Collapse
Affiliation(s)
- Samuel M Lancaster
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Brittany Lee-McMullen
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Charles Wilbur Abbott
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Jeniffer V Quijada
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Daniel Hornburg
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Heyjun Park
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Dalia Perelman
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Dylan J Peterson
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Michael Tang
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Aaron Robinson
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Sara Ahadi
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Chia-Jui Hung
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Melanie Ashland
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Tracey McLaughlin
- Division of Endocrinology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Anna Boonyanit
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Aaron Horning
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Justin L Sonnenburg
- Department of Microbiology & Immunology, Stanford School of Medicine, Stanford, CA 94305, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Michael P Snyder
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
36
|
Matsuoka T, Hosomi K, Park J, Goto Y, Nishimura M, Maruyama S, Murakami H, Konishi K, Miyachi M, Kawashima H, Mizuguchi K, Kobayashi T, Yokomichi H, Kunisawa J, Yamagata Z. Relationships between barley consumption and gut microbiome characteristics in a healthy Japanese population: a cross-sectional study. BMC Nutr 2022; 8:23. [PMID: 35287729 PMCID: PMC8919566 DOI: 10.1186/s40795-022-00500-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/03/2022] [Indexed: 11/29/2022] Open
Abstract
Background Barley contains abundant soluble beta-glucan fibers, which have established health benefits. In addition, the health benefits conferred by the gut bacteria have attracted considerable interest. However, few studies have focused on the barley consumption and gut bacteria of the Japanese population. In this study, we aimed to identify the relationship between the barley consumption and gut bacteria composition of the Japanese population. Methods In total, 236 participants were recruited in Japan, and 94 participants with no complications of diabetes, hypertension, or dyslipidemia were selected for the study. We analyzed fecal samples from the participants, their medical check-up results, and responses to questionnaires about dietary habits. The participants were grouped according to their median barley consumption. Then, we assessed the relative abundance of 50 genera. Characteristic bacteria were evaluated for their relationship with barley consumption by multiple regression analysis, adjusted for disease and dietary habits, in all participants. We also analyzed the networks and clustering of the 20 selected genera. Results According to the comparison between barley groups, Bifidobacterium, Butyricicoccus, Collinsella, Ruminococcus 2, and Dialister were characteristic candidate bacterias of the group that consumed large amounts of barley (P < 0.05). The relationship between barley consumption and Bifidobacterium remained after adjusting for disease and dietary habits, and that of Butyricicoccus remained after adjusting for disease. Furthermore, network and cluster analyses revealed that barley consumption was directly correlated with Bifidobacterium and Butyricicoccus. Conclusions Barley consumption generates changes in the intestinal bacteria of the Japanese population. We found that Bifidobacterium and Butyricicoccus abundance was positively associated with barley consumption. Supplementary Information The online version contains supplementary material available at 10.1186/s40795-022-00500-3.
Collapse
|
37
|
Comprehensive analysis of Sparassis crispa polysaccharide characteristics during the in vitro digestion and fermentation model. Food Res Int 2022; 154:111005. [DOI: 10.1016/j.foodres.2022.111005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 01/21/2022] [Accepted: 02/10/2022] [Indexed: 12/20/2022]
|
38
|
ANGGELA, HARMAYANI E, SETYANINGSIH W, WICHIENCHOT S. Prebiotic effect of porang oligo-glucomannan using fecal batch culture fermentation. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.06321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- ANGGELA
- Universitas Gadjah Mada, Indonesia; Prince of Songkla University, Thailand
| | | | | | | |
Collapse
|
39
|
Abstract
The neonatal body provides a range of potential habitats, such as the gut, for microbes. These sites eventually harbor microbial communities (microbiotas). A "complete" (adult) gut microbiota is not acquired by the neonate immediately after birth. Rather, the exclusive, milk-based nutrition of the infant encourages the assemblage of a gut microbiota of low diversity, usually dominated by bifidobacterial species. The maternal fecal microbiota is an important source of bacterial species that colonize the gut of infants, at least in the short-term. However, development of the microbiota is influenced by the use of human milk (breast feeding), infant formula, preterm delivery of infants, caesarean delivery, antibiotic administration, family details and other environmental factors. Following the introduction of weaning (complementary) foods, the gut microbiota develops in complexity due to the availability of a diversity of plant glycans in fruits and vegetables. These glycans provide growth substrates for the bacterial families (such as members of the Ruminococcaceae and Lachnospiraceae) that, in due course, will dominate the gut microbiota of the adult. Although current data are often fragmentary and observational, it can be concluded that the nutrition that a child receives in early life is likely to impinge not only on the development of the microbiota at that time but also on the subsequent lifelong, functional relationships between the microbiota and the human host. The purpose of this review, therefore, is to discuss the importance of promoting the assemblage of functionally robust gut microbiotas at appropriate times in early life.
Collapse
Affiliation(s)
- Gerald W. Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
40
|
Li D, Shu G, Wang H, Xu Y, Adni J, Zhang Y, MacAdam JW, Villalba JJ, Dai X, Chen L. In vitro fermentation performance of alfalfa (Medicago sativa L.) mixed with different proportions of paper mulberry (Broussonetia papyrifera) leaves (PML) or condensed tannins extracted from PML. ITALIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1080/1828051x.2021.1990144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Daojie Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Gangqin Shu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Hui Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Yuan Xu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Jannati Adni
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Yunhua Zhang
- School of Resources and Environment, Anhui Agricultural University, Hefei, China
| | - Jennifer W. MacAdam
- Department of Plants, Soils & Climate, Utah State University, Logan, UT, USA
| | - Juan J. Villalba
- Department of Wildland Resources, Utah State University, Logan, UT, USA
| | - Xin Dai
- Department of Plants, Soils & Climate, Utah State University, Logan, UT, USA
| | - Lijuan Chen
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| |
Collapse
|
41
|
Zhang S, Waterhouse GIN, Xu F, He Z, Du Y, Lian Y, Wu P, Sun-Waterhouse D. Recent advances in utilization of pectins in biomedical applications: a review focusing on molecular structure-directing health-promoting properties. Crit Rev Food Sci Nutr 2021:1-34. [PMID: 34637646 DOI: 10.1080/10408398.2021.1988897] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The numerous health benefits of pectins justify their inclusion in human diets and biomedical products. This review provides an overview of pectin extraction and modification methods, their physico-chemical characteristics, health-promoting properties, and pharmaceutical/biomedical applications. Pectins, as readily available and versatile biomolecules, can be tailored to possess specific functionalities for food, pharmaceutical and biomedical applications, through judicious selection of appropriate extraction and modification technologies/processes based on green chemistry principles. Pectin's structural and physicochemical characteristics dictate their effects on digestion and bioavailability of nutrients, as well as health-promoting properties including anticancer, immunomodulatory, anti-inflammatory, intestinal microflora-regulating, immune barrier-strengthening, hypercholesterolemia-/arteriosclerosis-preventing, anti-diabetic, anti-obesity, antitussive, analgesic, anticoagulant, and wound healing effects. HG, RG-I, RG-II, molecular weight, side chain pattern, and degrees of methylation, acetylation, amidation and branching are critical structural elements responsible for optimizing these health benefits. The physicochemical characteristics, health functionalities, biocompatibility and biodegradability of pectins enable the construction of pectin-based composites with distinct properties for targeted applications in bioactive/drug delivery, edible films/coatings, nano-/micro-encapsulation, wound dressings and biological tissue engineering. Achieving beneficial synergies among the green extraction and modification processes during pectin production, and between pectin and other composite components in biomedical products, should be key foci for future research.
Collapse
Affiliation(s)
- Shikai Zhang
- College of Food Science and Engineering, Shandong Agricultural University, Taian, China
| | | | - Fangzhou Xu
- College of Food Science and Engineering, Shandong Agricultural University, Taian, China
| | - Ziyang He
- College of Food Science and Engineering, Shandong Agricultural University, Taian, China
| | - Yuyi Du
- College of Food Science and Engineering, Shandong Agricultural University, Taian, China
| | - Yujing Lian
- College of Food Science and Engineering, Shandong Agricultural University, Taian, China
| | - Peng Wu
- College of Food Science and Engineering, Shandong Agricultural University, Taian, China
| | - Dongxiao Sun-Waterhouse
- College of Food Science and Engineering, Shandong Agricultural University, Taian, China.,School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
42
|
Laursen MF. Gut Microbiota Development: Influence of Diet from Infancy to Toddlerhood. ANNALS OF NUTRITION & METABOLISM 2021; 77:1-14. [PMID: 34461613 DOI: 10.1159/000517912] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/15/2021] [Indexed: 11/19/2022]
Abstract
Early life is a critical period as our gut microbiota establishes here and may impact both current and future health. Thus, it is of importance to understand how different factors govern the complex microbial colonization patterns in this period. The gut microbiota changes substantially during infancy and toddlerhood in terms of both taxonomic composition and diversity. This developmental trajectory differs by a variety of factors, including term of birth, mode of birth, intake of antibiotics, presence of furred pets, siblings and family members, host genetics, local environment, geographical location, and maternal and infant/toddler diet. The type of milk feeding and complementary feeding is particularly important in early and late infancy/toddlerhood, respectively. Breastfeeding, due to the supply of human milk oligosaccharide into the gut, promotes the growth of specific human milk oligosaccharide (HMO)-utilizing Bifidobacterium species that dominate the ecosystem as long as the infant is primarily breastfed. These species perform saccharolytic fermentation in the gut and produce metabolites with physiological effects that may contribute to protection against infectious and immune-related diseases. Formula feeding, due to its lack of HMOs and higher protein content, give rise to a more diverse gut microbiota that contains more opportunistic pathogens and results in a more proteolytic metabolism in the gut. Complementary feeding, due to the introduction of dietary fibers and new protein sources, induces a shift in the gut microbiota and metabolism away from the milk-adapted and toward a more mature and diverse adult-like community with increased abundances of short chain fatty acid-producing bacterial taxa. While the physiological implication of these complementary diet-induced changes remains to be established, a few recent studies indicate that an inadequately matured gut microbiota may be causally related to poor growth and development. Further studies are required to expand our knowledge on interactions between diet, gut microbiota, and health in the early life setting.
Collapse
|
43
|
Sauvaitre T, Etienne-Mesmin L, Sivignon A, Mosoni P, Courtin CM, Van de Wiele T, Blanquet-Diot S. Tripartite relationship between gut microbiota, intestinal mucus and dietary fibers: towards preventive strategies against enteric infections. FEMS Microbiol Rev 2021; 45:5918835. [PMID: 33026073 DOI: 10.1093/femsre/fuaa052] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
The human gut is inhabited by a large variety of microorganims involved in many physiological processes and collectively referred as to gut microbiota. Disrupted microbiome has been associated with negative health outcomes and especially could promote the onset of enteric infections. To sustain their growth and persistence within the human digestive tract, gut microbes and enteric pathogens rely on two main polysaccharide compartments, namely dietary fibers and mucus carbohydrates. Several evidences suggest that the three-way relationship between gut microbiota, dietary fibers and mucus layer could unravel the capacity of enteric pathogens to colonise the human digestive tract and ultimately lead to infection. The review starts by shedding light on similarities and differences between dietary fibers and mucus carbohydrates structures and functions. Next, we provide an overview of the interactions of these two components with the third partner, namely, the gut microbiota, under health and disease situations. The review will then provide insights into the relevance of using dietary fibers interventions to prevent enteric infections with a focus on gut microbial imbalance and impaired-mucus integrity. Facing the numerous challenges in studying microbiota-pathogen-dietary fiber-mucus interactions, we lastly describe the characteristics and potentialities of currently available in vitro models of the human gut.
Collapse
Affiliation(s)
- Thomas Sauvaitre
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France.,Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| | - Adeline Sivignon
- Université Clermont Auvergne, UMR 1071 Inserm, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH), Clermont-Ferrand, France
| | - Pascale Mosoni
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| | - Christophe M Courtin
- KU Leuven, Faculty of Bioscience Engineering, Laboratory of Food Chemistry and Biochemistry & Leuven Food Science and Nutrition Research Centre (LFoRCe), Leuven, Belgium
| | - Tom Van de Wiele
- Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| |
Collapse
|
44
|
Lee S, Goodson ML, Vang W, Rutkowsky J, Kalanetra K, Bhattacharya M, Barile D, Raybould HE. Human milk oligosaccharide 2'-fucosyllactose supplementation improves gut barrier function and signaling in the vagal afferent pathway in mice. Food Funct 2021; 12:8507-8521. [PMID: 34308934 PMCID: PMC8451585 DOI: 10.1039/d1fo00658d] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
2′-Fucosyllactose (2′-FL) is one of the predominant oligosaccharides found in human milk and has several well-established beneficial effects in the host. It has previously been shown that 2′-FL can improve the metabolic phenotype in high-fat (HF)-fed mice. Here we investigated whether dietary supplementation with 2′-FL was associated with improved intestinal barrier integrity, signaling in the vagal afferent pathway and cognitive function. Mice were fed either a low-fat (LF, 10% fat per kcal) or HF (45% fat per kcal) diet with or without supplementation of 2′-FL (10% w/w) in the diet for 8 weeks. Body weight, energy intake, fat and lean mass, intestinal permeability (ex vivo in Ussing chambers), lipid profiles, gut microbiome and microbial metabolites, and cognitive functions were measured. Vagal afferent activity was measured via immunohistochemical detection of c-Fos protein in the brainstem in response to peripheral administration of cholecystokinin (CCK). 2′-FL significantly attenuated the HF-induced increase in fat mass and energy intake. 2′-FL significantly reduced intestinal permeability and significantly increased expression of interleukin (IL)-22, a cytokine known for its protective role in the intestine. Additionally, 2′-FL led to changes in the gut microbiota composition and in the associated microbial metabolites. Signaling in the vagal afferent pathway was improved but there was no effect on cognitive function. In conclusion, 2′-FL supplementation improved the metabolic profiles, gut barrier integrity, lipid metabolism and signaling in the vagal afferent pathway. These findings support the utility of 2′-FL in the control of gut barrier function and metabolic homeostasis under a metabolic challenge. 2’-Fucosyllactose (2’-FL), a predominant human milk oligosaccharide, attenuates HF diet-induced metabolic and intestinal barrier impairment, improves gut hormone resistance, and alters the intestinal microbiota and microbiota-derived metabolites.![]()
Collapse
Affiliation(s)
- Sunhye Lee
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, UC Davis, CA, USA.
| | - Michael L Goodson
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, UC Davis, CA, USA.
| | - Wendie Vang
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, UC Davis, CA, USA.
| | - Jennifer Rutkowsky
- Department of Molecular Biosciences, School of Veterinary Medicine, UC Davis, CA, USA
| | - Karen Kalanetra
- Department of Food Science and Technology, College of Agriculture, UC Davis, CA, USA
| | - Mrittika Bhattacharya
- Department of Food Science and Technology, College of Agriculture, UC Davis, CA, USA
| | - Daniela Barile
- Department of Food Science and Technology, College of Agriculture, UC Davis, CA, USA
| | - Helen E Raybould
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, UC Davis, CA, USA.
| |
Collapse
|
45
|
A review on enzyme-producing lactobacilli associated with the human digestive process: From metabolism to application. Enzyme Microb Technol 2021; 149:109836. [PMID: 34311881 DOI: 10.1016/j.enzmictec.2021.109836] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022]
Abstract
Complex carbohydrates, proteins, and other food components require a longer digestion process to be absorbed by the lining of the alimentary canal. In addition to the enzymes of the gastrointestinal tract, gut microbiota, comprising a large range of bacteria and fungi, has complementary action on the production of digestive enzymes. Within this universe of "hidden soldiers", lactobacilli are extensively studied because of their ability to produce lactase, proteases, peptidases, fructanases, amylases, bile salt hydrolases, phytases, and esterases. The administration of living lactobacilli cells has been shown to increase nutrient digestibility. However, it is still little known how these microbial-derived enzymes act in the human body. Enzyme secretion may be affected by variations in temperature, pH, and other extreme conditions faced by the bacterial cells in the human body. Besides, lactobacilli administration cannot itself be considered the only factor interfering with enzyme secretion, human diet (microbial substrate) being determinant in their metabolism. This review highlights the potential of lactobacilli to release functional enzymes associated with the digestive process and how this complex metabolism can be explored to contribute to the human diet. Enzymatic activity of lactobacilli is exerted in a strain-dependent manner, i.e., within the same lactobacilli species, there are different enzyme contents, leading to a large variety of enzymatic activities. Thus, we report current methods to select the most promising lactobacilli strains as sources of bioactive enzymes. Finally, a patent landscape and commercial products are described to provide the state of art of the transfer of knowledge from the scientific sphere to the industrial application.
Collapse
|
46
|
Herman A, Bajaka A. The role of the intestinal microbiota in eating disorders - bulimia nervosa and binge eating disorder. Psychiatry Res 2021; 300:113923. [PMID: 33857846 DOI: 10.1016/j.psychres.2021.113923] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 04/01/2021] [Indexed: 02/08/2023]
Abstract
Bulimia nervosa (BN) and binge eating disorder (BED) are both eating disorders (EDs) characterised by episodes of overeating in which large amounts of food are consumed in short periods. The aetiology of BN and BED is not fully understood. Psychological and social factors influence the development of BN and BED, but biological factors such as neurohormones that regulate hunger and satiety, or neurotransmitters responsible for mood and anxiety play a significant role in sustaining symptoms. Increasing numbers of studies confirm the relationship between the composition of intestinal microbiota and the regulation of appetite, mood, and body mass. In this manuscript, we will describe the mechanisms by which intestinal dysbiosis can play an important role in the aetiology of binge eating episodes based on current understanding. Understanding the two-way relationship between BN and BED and alterations in the intestinal microbiota suggest the utility of new treatment methods of these disorders aimed at improving the composition of the intestinal microflora.
Collapse
Affiliation(s)
- Anna Herman
- Department of Child Psychiatry, Medical University of Warsaw, 61 Żwirki i Wigury St., 02-091 Warsaw, Poland.
| | - Armand Bajaka
- Department of Child Psychiatry, Medical University of Warsaw, 61 Żwirki i Wigury St., 02-091 Warsaw, Poland
| |
Collapse
|
47
|
Trefflich I, Dietrich S, Braune A, Abraham K, Weikert C. Short- and Branched-Chain Fatty Acids as Fecal Markers for Microbiota Activity in Vegans and Omnivores. Nutrients 2021; 13:1808. [PMID: 34073495 PMCID: PMC8230270 DOI: 10.3390/nu13061808] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/18/2021] [Accepted: 05/21/2021] [Indexed: 12/15/2022] Open
Abstract
A vegan diet could impact microbiota composition and bacterial metabolites like short-chain (SCFA) and branched-chain fatty acids (BCFA). The aim of this study was to compare the concentrations of SCFA, BCFA, ammonia, and fecal pH between vegans and omnivores. In this cross-sectional study (vegans n = 36; omnivores n = 36), microbiota composition, fecal SCFA, BCFA, and ammonia concentrations and pH were analyzed in complete stool samples. A random forest regression (RFR) was used to identify bacteria predicting SCFA/BCFA concentrations in vegans and omnivores. No significant differences in SCFA and BCFA concentrations were observed between vegans and omnivores. Fecal pH (p = 0.005) and ammonia concentration (p = 0.01) were significantly lower in vegans than in omnivores, while fiber intake was higher (p < 0.0001). Shannon diversity was higher in omnivores compared to vegans on species level (p = 0.04) only. In vegans, a cluster of Faecalibacterium prausnitzii, Prevotella copri, Dialister spp., and Eubacterium spp. was predictive for SCFA and BCFA concentrations. In omnivores, Bacteroides spp., Clostridium spp., Ruminococcus spp., and Prevotella copri were predictive. Though SCFA and BCFA did not differ between vegans and omnivores, the results of the RFR suggest that bacterial functionality may be adapted to varying nutrient availability in these diets.
Collapse
Affiliation(s)
- Iris Trefflich
- Department of Food Safety, German Federal Institute for Risk Assessment, 10589 Berlin, Germany; (S.D.); (K.A.); (C.W.)
| | - Stefan Dietrich
- Department of Food Safety, German Federal Institute for Risk Assessment, 10589 Berlin, Germany; (S.D.); (K.A.); (C.W.)
| | - Annett Braune
- Research Group Intestinal Microbiology, Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558 Nuthetal, Germany;
| | - Klaus Abraham
- Department of Food Safety, German Federal Institute for Risk Assessment, 10589 Berlin, Germany; (S.D.); (K.A.); (C.W.)
| | - Cornelia Weikert
- Department of Food Safety, German Federal Institute for Risk Assessment, 10589 Berlin, Germany; (S.D.); (K.A.); (C.W.)
| |
Collapse
|
48
|
Su Q, Liu Q. Factors Affecting Gut Microbiome in Daily Diet. Front Nutr 2021; 8:644138. [PMID: 34041257 PMCID: PMC8141808 DOI: 10.3389/fnut.2021.644138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 04/16/2021] [Indexed: 12/13/2022] Open
Abstract
There is a growing recognition that a good diet can help people maintain mental and physical health, while a bad one will cause the disorder of body function, and even lead to several diseases. A lot of attentions have been devoted to analyze every possible health-related factor in the daily diet, including food ingredients, additives, and cooking process. With the support of high-throughput sequencing technology, there is accumulating evidence gradually clarifying that most of these factors are mainly through the interactions with gut microbiome to trigger downstream effects. The gut microbiome may be able to act as a very sensitive mirror in response to human daily diet. A complex network of interactions among diet, gut microbiome, and health has been gradually depicted, but it is rarely discussed from a more comprehensive perspective. To this end, this review summarized the latest updates in diet-gut microbiome interactions, analyzed most identified factors involved in this process, showed the possibility of maintaining health or alleviating diseases by diet intervention, aiming to help people choose a suitable recipe more accurately.
Collapse
Affiliation(s)
| | - Qin Liu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
49
|
Zhu L, Gao M, Li H, Deng ZY, Zhang B, Fan Y. Effects of soluble dietary fiber from sweet potato dregs on the structures of intestinal flora in mice. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.100880] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
50
|
Yang Z, Zhang C, Wang J, Celi P, Ding X, Bai S, Zeng Q, Mao X, Zhuo Y, Xu S, Yan H, Zhang K, Shan Z. Characterization of the Intestinal Microbiota of Broiler Breeders With Different Egg Laying Rate. Front Vet Sci 2020; 7:599337. [PMID: 33330722 PMCID: PMC7732610 DOI: 10.3389/fvets.2020.599337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/03/2020] [Indexed: 11/24/2022] Open
Abstract
The gastrointestinal microbiota plays a pivotal role in maintaining animal health, immunity and reproductive performances. However, literature about the relationship between microbiota and reproductive performance is limited. The aim of the present study was to determine differences in the intestinal microbiota of broiler breeders with different egg laying rate. A total of 200 AA+ parent broiler breeders (41-week-old) were separated into two groups according to their different egg laying rate [average egg laying rate group (AR: 78.57 ± 0.20%) and high egg laying rate group (HR: 90.79 ± 0.43%). Feed conversion ratio (FCR), ovary cell apoptosis rate (ApoCR) and relative abdominal fat weight were lower (p = 0.01), while the hatchability rate of qualified egg was higher (p = 0.04) in HR group than that in AR group. Phascolarctobacterium abundance were lower (p = 0.012) in ileum of HR birds. Romboutsia (genus) in ileum was negatively related to the feed efficiency (r = -0.58, p < 0.05), Firmicutes (phylum) and Lactobacillus (genus) abundances in cecum were positively related to the egg laying rate (ELR) (r = 0.35 and 0.48, p < 0.05), feed efficiency (r = 0.42 and 0.43, p < 0.05), while Spirochaetes (phylum) and Sphaerochaeta (genus) abundances in cecum were negatively related to the ELR (r = -0.43 and -0.70, p < 0.05), feed efficiency (r = 0.54 and 0.48, p < 0.05), and positively related to ApoCR (r = 0.46 and 0.47, p < 0.05). Our results suggested that microbiota, such as Firmicutes (phylum) and Lactobacillus (genus) have positive relationship, while Spirochaetes (phylum) and Romboutsia (genus) abundances exert negative relationship with broiler breeders' reproductive performances.
Collapse
Affiliation(s)
- Zengqiao Yang
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Chunhua Zhang
- College of Agriculture and Forestry, Pu'er University, Pu'er City, China
| | - Jianping Wang
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Pietro Celi
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Xuemei Ding
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Shiping Bai
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Qiufeng Zeng
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Xiangbing Mao
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Yong Zhuo
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Shengyu Xu
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Hui Yan
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Keying Zhang
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Zhiguo Shan
- College of Agriculture and Forestry, Pu'er University, Pu'er City, China
| |
Collapse
|